WO2002086083A2 - Procedes d'amelioration de la capacite de reaction de cellules t - Google Patents

Procedes d'amelioration de la capacite de reaction de cellules t Download PDF

Info

Publication number
WO2002086083A2
WO2002086083A2 PCT/US2002/012733 US0212733W WO02086083A2 WO 2002086083 A2 WO2002086083 A2 WO 2002086083A2 US 0212733 W US0212733 W US 0212733W WO 02086083 A2 WO02086083 A2 WO 02086083A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cancer
cell
human
tumor
Prior art date
Application number
PCT/US2002/012733
Other languages
English (en)
Other versions
WO2002086083A3 (fr
Inventor
Lieping Chen
Scott E. Strome
Original Assignee
Mayo Foundation For Medical Education And Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mayo Foundation For Medical Education And Research filed Critical Mayo Foundation For Medical Education And Research
Priority to AU2002258941A priority Critical patent/AU2002258941A1/en
Publication of WO2002086083A2 publication Critical patent/WO2002086083A2/fr
Publication of WO2002086083A3 publication Critical patent/WO2002086083A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4632T-cell receptors [TCR]; antibody T-cell receptor constructs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464484Cancer testis antigens, e.g. SSX, BAGE, GAGE or SAGE
    • A61K39/464486MAGE
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/51B7 molecules, e.g. CD80, CD86, CD28 (ligand), CD152 (ligand)

Definitions

  • TECHNICAL FIELD This invention relates to enhancing immunity, and more particularly to enhancing T cell responsiveness.
  • T lymphocytes are subject to various regulatory processes. It is important in some diseases (e.g., cancer) that imrnunosuppression mediated by disease-associated mechanisms be controlled so as to control progression of the disease.
  • diseases e.g., cancer
  • the invention is based on the discovery that B7-H1 molecules (e.g., on cancer cells) inhibit proliferation of T cells and/or apoptosis of activated T cells (CD4+ and CD8+) and thereby act as negative immunoregulatory molecules.
  • B7-Hl e.g., on cancer cells
  • apoptosis of activated T cells CD4+ and CD8+
  • Expression of B7- Hl by a variety of cancer cells represents a newly discovered mechanism whereby such cancer cells evade a host's immune system.
  • the inventor has also discovered that agents that inhibit the interaction between B7-H1 and T cells reverse the negative regulatory effect of B7-H1 on T cells and thus enhance T cell responsiveness.
  • the invention thus features methods of enhancing T cell responsiveness.
  • Human B7-H1 is designated hB7-Hl and mouse B7-H1 is designated mB7- Hl. Where the term B7-H1 without specifying either species is used, the relevant text applies to B7-H1 molecules of any species.
  • the invention provides a method of enhancing T cell responsiveness in a mammal.
  • the method includes the steps of: (a) identifying a mammal with, or at risk of developing, cancer, the cells of the cancer being identified as expressing B7-H1 molecules on their surfaces; and (b) administering to the subject l a compound that includes an agent that interferes with an interaction between B7-H1 and a T cell.
  • the method can further involve administering an immunogenic stimulus to the mammal.
  • the agent can be, for example, an antibody that binds to B7-H1, B7-H1 or a functional fragment of B7-H1, or a receptor for B7- HI or a functional fragment of a receptor for B7-H1.
  • the compound can be a fusion protein, e.g., a fusion protein that contains all, or part, of an immunoglobulin constant region.
  • the cancer can be, for example, a hematological cancer, a neurological cancer, melanoma, breast cancer, lung cancer, head and neck cancer, a gastrointestinal cancer, liver cancer, pancreatic cancer, a genitourinary cancer, a bone cancer, and a vascular cancer and the mammal can be, for example, a human.
  • the invention also embodies a method of enhancing the responsiveness of a T cell.
  • the method involves culturing, together, a T cell, a cancer cell identified as expressing B7-H1, and a compound comprising an agent that interferes with an interaction between B7-H1 and a T cell.
  • the cultures can optionally also include an immunogenic stimulus.
  • the agent can any of those listed above and the cancer cell can be of any of the cancers listed above.
  • the T cell can be a CD8+ T cell or a CD4+ T cell.
  • the compound can be a fusion protein, e.g., the fusion protein described above.
  • enhancing T cell responsiveness means enhancing the ability of a T cell to respond to an immunogenic stimulus.
  • the enhancement is not limited by any particular mechanism of action and can mean, for example, reversing (completely or partially) inhibition of the ability of the T cell to proliferate or by preventing the cell from dying by, for example, apoptosis or necrosis.
  • (b) modifies the physical interaction of the T cell with a B7-H1 molecule such that the physical interaction either does not deliver a signal to the T cell or delivers a signal to the T cell that does not substantially result in either death of the T cell or inhibition of the T cell's function, e.g., cytotoxic or helper activity.
  • a "functional fragment" of a B7-Hl polypeptide is a fragment of the B7-H1 polypeptide that is shorter than the full-length polypeptide and has the ability to bind to a T cell.
  • a functional fragment of a receptor for B7- Hl e.g., PD-1
  • PD-1 a fragment of the receptor for B7-H1 that has the ability to bind to B7-H1.
  • inhibiting proliferation of a cancer cell means either completely inhibiting proliferation of the cancer cell or substantially reducing the rate of proliferation of the cancer cell, e.g., reducing the rate of proliferation by 10%; 20%; 30%; 40%; 50%; 60%; 70%; 80%; 90%; 95%; 98%; 99%; or 99.5%; or 99.9%.
  • an antibody refers not only to whole antibody molecules, but also to antigen-binding fragments, e.g., Fab, F(ab') 2 , Fv, and single chain Fv (sFv) fragments.
  • An sFv fragment is a single polypeptide chain that includes both the heavy and light chain variable regions of the antibody from which the sFv is derived. Such fragments can be produced, for example, as described in U.S. Patent No. 4,642,334, which is incorporated herein by reference in its entirety. Also included are chimeric antibodies.
  • Chimeric antibodies are recombinant antibodies comprising portions derived from more than one species; for example, the antigen binding regions (i.e., the complementarity determining regions (CDR)), of the antibody molecule can be derived from a mouse wild-type antibody molecule and framework and constant regions can be derived from a human antibody molecule or human antibody molecules.
  • CDR complementarity determining regions
  • Fig. 1A is a line graph showing the level of T cell proliferation measured as counts per minute (cpm x 10 3 ) of 3 H-thymidine incorporated ( 3 H-TdR uptake) into T cells after 40, 64, and 88 hours of culture of purified human T cells (2 x 10 5 per well) in the presence of tissue culture plate-bound hB7-HHg fusion protein (or control Ig) (each coated onto the plates at a concentration of 10 ⁇ g/ml) and tissue culture-plate- bound antibody specific for human CD3 (coated onto the plates at a concentration of 500 ng/ml).
  • tissue culture plate-bound hB7-HHg fusion protein or control Ig
  • soluble PD-lIg fusion protein or control mouse IgG2a (mlgG2a)) (each at a concentration of 30 ⁇ g/ml).
  • Fig. IB is a line graph showing the concentration of viable cells (measured using the trypan blue exclusion assay) after 40, 64, and 88 hours of culture of purified human T cells (2 x 10 5 per well) in the presence of tissue culture plate-bound hB7- Hllg fusion protein (or control Ig) (each coated onto the plates at a concentration of 10 ⁇ g/ml) and tissue culture-plate-bound antibody specific for human CD3 (coated onto the plates at a concentration of 500 ng/ml).
  • tissue culture plate-bound hB7- Hllg fusion protein or control Ig
  • tissue culture-plate-bound antibody specific for human CD3 coated onto the plates at a concentration of 500 ng/ml.
  • Some culture wells soluble PD-1 Ig fusion protein or control mouse IgG2a (
  • Fig. 1C is a line graph showing the concentration of apoptotic cells (detected by fluorescence flow cytometry (FFC) as cells staining with Annexin V (AV+) and not staining with propidium iodide (PI-)) after 40, 64, and 88 hours of culture of purified human T cells (2 x 10 5 per well) in the presence of tissue culture plate-bound hB7-HHg fusion protein (or control Ig) (each coated onto the plates at a concentration of 10 ⁇ g/ml) and tissue culture-plate-bound antibody specific for human CD3 (coated onto the plates at a concentration of 500 ng/ml).
  • Some culture wells contained soluble PD-lIg fusion protein (or control mouse IgG2a (mlgG2a)) (each at a concentration of 30 ⁇ g/ml).
  • Fig. 2A is a pair of FFC histograms showing the expression of hB7-Hl (B7- Hl) and lack of expression of Fas ligand by cells of the NCI-H292 human lung carcinoma cell line.
  • the cells were stained with antibody specific for hB7-Hl (left histogram) or Fas ligand (right histogram) and fluorescein isothiocyanate- (FITC-) conjugated F(ab') 2 goat anti-mouse IgG antibody (filled profiles) or with FITC- conjugated F(ab') 2 goat anti-mouse IgG antibody only (unfilled profiles).
  • FITC- fluorescein isothiocyanate-
  • Fig. 2B is series of six two-dimensional FFC histograms of purified human T cells that had been cultured (2 x 10 5 per well) alone (T cells) or with irradiated NCT- H292 cells (4 x 10 4 per well) (T cells + H292) for 5 days and then stained with 7- AAD, anti-CD4, and anti-CD8 antibody.
  • the histograms on the right show the data in terms cells of staining with 7-AAD and forward light scatter (FSC) which is a function of cell size; the R2 (apoptotic cells) and R3 gates (live cells) are indicated.
  • the cells in R2 and R3 gates were then analyzed for their expression of CD4 and CD8 and the relevant data are shown in the four two-dimensional FFC histograms on the right.
  • Fig. 2C is a bar graph showing the % increase of apoptotic cells when 20 ⁇ g/ml of either soluble 2H11 mAb specific for hB7-Hl (anti-B7-Hl), soluble PD-lIg fusion protein (PD-lIg), soluble l ⁇ B7-HHg fusion protein (B7-Hllg), or soluble control mouse IgG were added to cultures containing purified human T cells and NCI-H292 cells (as described for Fig. 2A). The apoptotic cells were detected by staining with 7- AAD.
  • Fig. 3 A is a series of ten FFC histograms showing the expression (or lack of expression) of HLA-DR, HLA-DQ, B7-H1, Fas Ligand, and TRAIL by cells of the Skel-28 human melanoma cell line that had been cultured in medium alone (Medium) or in medium containing interferon-E (IFN-E). Cells stained with antibodies specific for the listed cell-surface molecules are indicated by filled profiles and those stained with a control antibody (mouse IgGl) are indicated by unfilled profiles. Figs.
  • 3B and C are four line graphs showing the number of viable cells (top graphs) and the % apoptotic cells (bottom graphs) recovered from cultures of IFN-E treated Skmel-28 cells ( 2 x 10 4 per well) and cells of the melanoma peptide-epitope specific 8G9 CD4+ T cell clone (Fig. 3B) (2 x 10 5 per well) or cells of the alloantigen-specific 4D16 CD4+ T cell clone (2 x 10 5 per well) after 6 days of culture.
  • the cultures also contained as blocking agents (at the indicated concentrations) either 2H11 mAb specific for hB7-Hl (anti-B7-Hl) or control mouse Igd (MlgGl).
  • Fig. 3D is a series of four FFC histograms showing the expression of hB7-Hl by cells of the 624mel human melanoma cell line that had been stably transfected with a control expression vector (left histograms) or an expression vector containing a cDNA sequence encoding hB7-Hl (right histograms).
  • the cells were stained with either a mAb specific for hB7-Hl (Anti-B7-Hl) (top histograms; filled profiles), PD- 1 Ig fusion protein (bottom histograms; filled profiles), or control antibody (mouse IgGl; unfilled profiles).
  • Anti-B7-Hl top histograms; filled profiles
  • PD- 1 Ig fusion protein bottom histograms; filled profiles
  • control antibody mouse IgGl; unfilled profiles.
  • Fig. 4A is a bar graph showing the % of apoptotic cells recovered after 7 days from cultures containing cells of the CD8+ CTL clone M15 specific for a human melanoma peptide epitope alone (Medium) or with 624mel cells at a T cell to tumor cell ratio of 2:1.
  • the 624mel cells used had been stably transfected with a control expression vector (Mock/624mel) or an expression vector containing a cDNA sequence encoding hB7-Hl (B7-Hl/624mel).
  • Fig. 4B is a pair of line graphs showing the % of specific apoptosis (top graph) and the number of viable cells (bottom graph) in M15 cells after culture for 5 days with B7-Hl/624mel cells in the presence of the indicated concentrations of control mouse Ig (Control), 5H1 mAb specific for l ⁇ B7-Hl (5H1 ). F(ab') 2 fragments of the 5H1 mAb (5H1 F(ab')2), or PD-l Ig fusion protein.
  • apoptosis was calculated as [(% apoptotic T cells in an experimental culture containing tumor cells and a blocking agent/% apoptotic cells in the control culture without tumor cells or blocking agent) - 1] x 100.
  • the apoptotic cells were detected by FFC as those staining with Annexin V and the viable cells were detected by trypan blue exclusion.
  • Fig. 5 A is a series of photomicrographs of Mock/624mel cells (top panels) or B7-Hl/624mel cells (bottom panels) that has been cultured for 5 days with medium only (top left panel), Ml 5 cells (top right panel), Ml 5 cells and control mouse IgGl (10 ⁇ g/ml; bottom left panel), or M15 cells and 2H1 mAb specific for hB7-Hl (10 ⁇ g/ml;bottom right panel).
  • the cultures initially contained 3 x 10 tumor cells per well and those cultures containing M15 cells contained ' the same number of M15 cells.
  • Fig. 5B is a line graph showing the number of viable Mock/624mel cells or
  • B7-Hl/624mel cells (detected by trypan blue exclusion) recovered after various times of culture in the presence of M15 cells only, M15 cells and control mouse IgGl (mlgGl), or 2H1 mAb. Culture conditions were as described for Fig. 5A. Asterisks indicated data points that are significantly different (PO.05) from corresponding data points obtained from cultures containing B7-Hl/624mel cells, M15 cells, and mlgGl .
  • Fig. 5C is a line graph showing the level of cytolysis (% specific lysis as measured in a standard 51 Cr release assay) of Mock/624mel cells and B7-Hl/624mel cells after incubation with Ml 5 cells for 4 hours at the indicated Ml 5 cell to tumor target cell ratios (E/T).
  • E/T Ml 5 cell to tumor target cell ratios
  • 6A is a series of four FFC histograms showing: the expression by cells of the human breast carcinoma cell line HBL-100 of hB7-Hl as detected by staining with the 5H1 mAb specific for hB7-Hl (top left histogram) and the PD-l Ig fusion protein (top right histogram); and the lack of expression of Fas ligand and TRAIL as detected by staining with mAbs specific for the two cell surface markers (Anti-FasL and Anti-TRAIL, respectively). Fluorescence obtained by staining with the test reagents is shown by the filled profiles and staining obtained with control mAb is shown by unfilled profiles.
  • Fig. 6B is a pair of line graphs showing the % of apoptotic cells in the CD8+ cells (top graph) and the concentration of viable cells (bottom graph) in T cells recovered from cultures of HBL-100 cells and cells of the M99 CD8+ CTL clone M99 that is specific for a carcinoembryonic antigen (CEA) peptide epitope.
  • the cultures also contained either no blocking agent (Control), the 5H1 mAb specific for hB7-Hl (5H1) or the PD-lIg fusion protein (PD-lIg).
  • Apoptotic CD8+ cells and viable cells in recovered T cells were detected by FFC after staining with Annexin V and antibody specific for human CD8 and FFC and by trypan blue exclusion, respectively.
  • Fig. 7A is a line graph showing the % of apoptotic cells in cells recovered at various times from cultures of purified human T cells (2 x 10 per well) and plate bound hB7-Hg fusion protein (or control Ig) each coated onto the tissue culture plates at a concentration of 10 ⁇ g/ml and plate bound mAb specific for human CD3 coated onto the plates at an optimal dose
  • Fig. 7B is a series of four FFC histograms showing the expression of Fas ligand (top histograms) and Fas (bottom histograms) on cells recovered at 72 hours from the cultures described for Fig. 7A. Cells from only the cultures without blocking agents were tested.
  • Cells stained with a FITC-conjugated mAb specific for Fas ligand (bottom histograms) or a mAb specific for Fas ligand and FITC-conjugated goat F(ab') 2 anti-mouse IgG (top histograms) are indicated by the filled profiles and those stained with control antibodies are indicated by unfilled profiles.
  • the bars indicate the gating used to estimate the percentages % of cells expressing either Fas ligand or Fas and these percentages are shown above the bars.
  • Fig. 7C is a bar graph showing the % of apoptotic cells in cells recovered at 72 hours from the cultures described for Fig. 7A.
  • some of the cultures of the present experiment contained as blocking agents soluble neutralizing mAb specific for either interleukin-10 (Anti- TL10) or Fas ligand (Anti-Fas ligand) or a control antibody (Control Ab).
  • Apoptotic cells were detected as described for the experiment shown in Fig. 7A.
  • Figs. 8 A and B are line graphs showing the % of cells expressing the 2C transgenic T cell receptor (TCR) (TB2 + cells; Fig.
  • TCR 2C transgenic T cell receptor
  • Fig. 8B peritoneal cells recovered from P815 tumor-bearing RAG-1 " " imrriunodeficient mice at the indicated time points after i.p. injection with preactivated T cells (expressing the 2C transgenic TCR) (2.5 x 10 6 per mouse) from mice transgenic for the TCR specific peptide expressed by P815 tumor cells.
  • the preactivated T cells were prepared by culturing T cells purified from the spleens and lymph nodes of the 2C transgenic mice with irradiated spleen cells from BALB/c mice in the presence of human interleukin-2 (10 U/ml) for 3 days. The T cells were harvested from the cultures and depleted of dead cells by centrifugation over
  • the tumor cells (1 x 10 5 per mouse) injected i.p. into the RAG-l " " mice three days before injection of the preactivated T cells were P815 cells stably transfected with a either a control expression vector (Mock P815) or an expression vector containing cDNA fragment encoding mB7-Hl (B7-H1/P815).
  • the cells expressing the 2C transgenic receptor were detected by FFC after staining with a phycoerythrin-conjugated mAb (1B2) specific for the 2C TCR and apoptotic cells were detected by FFC after staining with FITC-labeled Annexin V.
  • B7-H1 enhanced the proliferation of T cells in response to suboptimal T cell receptor (TCR) activation in short term cultures, it inhibited the proliferation of the T cells in long-term cultures.
  • TCR suboptimal T cell receptor
  • B7-H1 enhanced apoptosis of the T cells.
  • the inventor also found that a wide variety of human tumor cells either constitutively express hB7-Hl on their surfaces or express it on their surfaces after exposure to interferon-E (IFN-E). While most normal tissues (e.g., breast, colon, pancreas, kidney, uterus, skeletal muscle, lung, liver, tonsil, and intestine) do not express hB7-Hl, it was found on at least some macrophages.
  • IFN-E interferon-E
  • the cells of a hB7-Hl -expressing human mucoepidermoid lung carcinoma cell line when cultured with peripheral blood T cells, caused enhanced apoptosis of the T cells, with the majority of apoptotic T cells being CD4+.
  • This enhanced apoptosis was inhibited by a monoclonal antibody (mAb) specific for hB7-Hl and by a soluble fusion protein containing hB7-Hl.
  • mAb monoclonal antibody
  • melanoma line (624mel) that do not constitutively express hB7-Hl were transfected with an expression vector so as to express cell surface hB7- HI.
  • the resulting recombinant melanoma cells were found to enhance apoptosis of human T cells in a manner similar to the cells of the lung carcinoma line.
  • Cells of a second human melanoma line (Skmel-28) treated with interferon E (IFN-E) were found to enhance the apoptosis of the cells of a CD4+ T cell clone specific for a melanoma peptide epitope and the enhanced apoptosis was inhibited by a Ab specific for hB7-Hl.
  • IFN-E interferon E
  • the recombinant 624mel melanoma cells also induced apoptosis in a significantly higher number of cells of a CD8+ cytotoxic T cell (CTL) clone specific for a melanoma peptide epitope than did mock-transfected melanoma cells.
  • CTL cytotoxic T cell
  • This increased apoptosis was inhibited by both a mAb specific for l B7-Hl and by a fusion protein (PD-lIg) containing the putative receptor (PD-1) for hB7-Hl and a part of the constant region of mouse IgG2a.
  • cells of a human breast cancer cell line that constitutively express hB7-Hl on their surfaces were found to induce apoptosis in cells of a CTL clone specific for a peptide epitope produced by the human breast cancer cells.
  • the apoptosis of these CTL was reduced by inclusion in the relevant of a mAb specific for B7-H1 in the culture medium.
  • the methods of the invention involve enhancing the responsiveness of a T cell. These methods are based on the findings that the co-stimulatory B7-H 1 molecule decreases proliferation and/or apoptosis of T cells and that agents that interfere with the physical interaction between B7-H1 a T cell reverse, completely or partially, this inhibitory effect. Thus the methods of the invention involve enhancing the responsiveness of T cells using compounds that contain agents that interfere with the physical interaction between B7-H1 and a T cell.
  • Cancer cells can be breast cancer, lung cancer, colon cancer, pancreatic cancer, renal cancer- stomach cancer, liver cancer, bone cancer, hematological cancer (e.g., leukemia or lymphoma), neural tissue cancer, melanoma, ovarian cancer, testicular cancer, prostate cancer, cervical cancer, vaginal cancer, bladder cancer cells or any other cancer cells listed herein.
  • the cells of the relevant cancer can optionally be tested for expression of B7-H1 by methods known in the art such as those disclosed herein.
  • Cells can be tested for expression of a polypeptide by, for example, fluorescence flow cytometry, fluorescence microscopy, or immunohistochemistry.
  • lysates of cells or culture supernatants of cells can be tested for the presence of the polypeptide of interest by electrophoretic methods optionally combined with immunoblotting methods.
  • immunoblotting can be performed without electrophoresis.
  • Lysates and culture supernatants of cells can also be tested by methods such as ELISA.
  • RNA in or isolated from the cells can also be analyzed for the presence of mRNA transcribed from the gene encoding the protein by, for example, in situ hybridization, spot blotting, Northern blotting, or reverse transcriptase-polymerase chain reaction (RT-PCR) analysis.
  • body fluids e.g. blood or urine
  • a subject e.g., a human cancer patient
  • body fluids e.g. blood or urine
  • a subject e.g., a human cancer patient
  • appropriately labeled (e.g., radiolabeled or fluorescently labeled) antibodies specific for B7-H1 can be used as agents to detect expression of B7-H1 by a cancer using established in vivo imaging (e.g., radiological) techniques.
  • a T cell whose responsiveness is inhibited by cancer cells.
  • the methods of the invention can be applied to enhancing the responsiveness of a T cell that is exposed to any cell that expresses B7-H1 and for which the T cell, by virtue of the antigen specific T cell receptor (TCR) it expresses, has specific responsiveness.
  • a cell can be, for example, an APC (e.g., a dendritic cell, a macrophage, a monocyte, or a B lymphocyte) that has a major histocompatibility complex (MHC) (class I or class II) molecule-peptide epitope molecular complex on its surface for which a relevant T cell is restricted and specific.
  • MHC major histocompatibility complex
  • Appropriate peptide epitopes include those derived from any of a variety of cancer cells (e.g., any of those listed herein) or infectious microorganisms (see below). Since cancer cell-derived peptide epitopes can be presented to T cells of interest by B7-H1 -expressing APC, the methods of the invention can be applied to treatment of cancers other than those expressing cell- surface B7-H1.
  • Peptide epitopes can also be derived from any of a variety of infectious microorganisms. It is understood that relevant cells need not be classical APC but can be any B7-H1 -expressing cells (or cells in which B7-H1 expression is inducible by, for example, IFN-E) infected with an appropriate infectious microorganism. Such cells include, without limitation, T cells, tissue epithelial cells, endothelial cells, and fibroblasts. Thus, the methods of the invention can be applied to the treatment of infections by any of a wide variety of infectious microorganisms.
  • microorganisms will generally be those that replicate inside a cell (commonly designated intracellular pathogens), since APC presenting peptide epitopes derived from infectious microorganism that are not necessarily in the relevant APC, the methods of the invention can also be applied to situations involving infectious microorganisms that replicate extracellularly or in cells that do not express B7-H1.
  • Relevant microorganisms can be viruses, bacteria, mycoplasma, fungi (including yeasts), and protozoan parasites and specific examples of such microorganisms include, without limitation, Mycohacteria tuberculosis, Salmonella enteriditis, Listeria monocytogenes, M.
  • Cryptococcus neoformans Chlamydia trachomatis, Candida albicans, Plasmodium falciparum, Entamoeba histolytica, Toxoplasma brucei, Toxoplasma gondii, Leishmania major, human immunodeficiency virus 1 and 2, influenza vims, measles virus, rabies virus, hepatitis vims A, B, and C, rotaviruses, papilloma virus, respiratory syncytial vims, feline immunodeficiency virus, feline leukemia vims, and simian immunodeficiency vims.
  • Compounds that can be used to enhance the responsiveness of a T cell can be, or contain agents, that interfere with the interaction between a T cell and B7-H1 or functional fragments of such agents. These compounds are referred to herein as "enhancing compounds.”
  • Appropriate agents include antibodies that bind to B7-H1, cell-free, soluble forms of receptors for B7-H 1 (e.g., PD1), or cell-free, soluble forms of B7-H1.
  • Such agents or functional fragments can have amino acid sequences identical to wild-type sequences or they can contain no more than 100 (e.g., no more than: two; three; four; five; six; seven; eight; nine; 10; 12; 14; 17; 20; 25; 30; 35; 40; 50; 60; 70; 80; 90; or 100 ) conservative amino acid substitutions.
  • an "immunogenic stimulus” is a stimulus delivered to a T cell via the antigen-specific T cell receptor (TCR) expressed on the surface of the T cell. More commonly, but not necessarily, such a stimulus is provided in the form of an antigen for which the TCR is specific. While such antigens will generally be protein, they can also be carbohydrates, lipids, nucleic acids or hybrid molecules having components of two or more of these molecule types, e.g., glycoproteins or lipoproteins.
  • TCR antigen-specific T cell receptor
  • the immunogenic stimulus can also be provided by other agonistic TCR ligands such as antibodies specific for TCR components (e.g., TCR E- chain or e-chain variable regions) or antibodies specific for the TCR-associated CD3 complex.
  • Immunogenic stimuli do not include antigen-non-specific stimuli provided by non-specifically acting factors such as, for example, cytokines (e.g., e.g., interleukin- (1L-)1, IL-2, IL-3, IL-4, IL-5, IL-6, 1L-10, TL-12, 1L-13, 1L-15, an interferon (JFN; e.g., IFN-E, TFN-e, IFN-E), granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF) or tumor necrosis factor-E (TNF-E)), co-stimulatory molecules, or adhesion molecules.
  • cytokines
  • Antigens useful as immunogenic stimuli include alloantigens (e.g., a MHC alloantigen) on, for example, an antigen presenting cell (APC) (e.g., a dendritic cell (DC), a macrophage, a monocyte, or a B cell).
  • APC antigen presenting cell
  • DC of interest are interdigitating DC and not follicular DC; follicular DC present antigen to B cells.
  • interdigitating DC are referred to herein as DC.
  • Methods of isolating DC from tissues such as blood, bone marrow, spleen, or lymph node are known in the art, as are methods of generating them in vitro from precursor cells in such tissues.
  • immunogenic stimuli are polypeptide antigens and peptide-epitopes derived from them (see below). Unprocessed polypeptides are processed by APC into peptide- epitopes that are presented to responsive T cells in the form of molecular complexes with MHC molecules on the surface of the APC.
  • Useful immunogenic stimuli also include a source of antigen such as a lysate of either tumor cells or cells infected with an infectious microorganism of interest.
  • APC pre-exposed (e.g., by coculturing) to antigenic polypeptides, peptide-epitopes of such polypeptides or lysates of tumor (or infected cells) can also be used as immunogenic stimuli.
  • Such APC can also be "primed” with antigen by culture with a cancer cell or infected cell of interest; the cancer or infected cells can optionally be irradiated or heated (e.g., boiled) prior to the priming culture.
  • APC especially DC
  • antigen as an immunogenic stimulus be provided in the form of cells (e.g., tumor cells or infected cells producing the antigen of interest).
  • immunogenic stimuli can be provided in the fonn of cell hybrids fom ed by fusing APC (e.g., DC) with tumor cells [Gong et al. (2000) Proc. Natl. Acad. Sci. USA 97(6):2716-2718; Gong et al. (1997) Nature Medicine 3(5):558-561 ; Gong et al. (2000) J. Immunol. 165(3): 1705- 371 1] or infected cells of interest.
  • APC e.g., DC
  • IC immunogenic cells
  • Cells or cell hybrids can be used (as immunogenic stimuli) untreated or they can be metabolically inhibited (e.g., by irradiation or exposure to a d g such as mitomycin-C) so as to substantially ablate their ability to divide.
  • Tumor or infected cells used per se as an immunogenic stimulus or as IC for the production of cell hybrids will preferably, but not necessarily, be derived from the same donor as that of the T cell.
  • the cells are from a different donor, they will preferably share one MHC haplotype with the T cell.
  • APC used to form cell hybrids will also preferably, but not necessarily, be derived from the same donor as the T cell. In the production of cell hybrids, either the APC or the IC will be preferably be from, or MHC-compatible with, the donor of the T cell. Alternatively, the APC and/or the IC can share one MHC haplotype (i.e., be semi-allogeneic) with the donor of the T cell.
  • the cells or hybrids used as immunogenic stimuli will frequently be used in the presence of APC of the T cell donor (e.g., in in vivo applications), they can be fully MHC incompatible with the T cell.
  • heat shock proteins bound to antigenic peptide-epitopes derived from antigens e.g., tumor-associated antigens or antigens produced by infectious microorganisms
  • antigens e.g., tumor-associated antigens or antigens produced by infectious microorganisms
  • Such complexes of heat shock protein and antigenic peptide are useful for facilitating or enhancing uptake of antigenic peptides by APC.
  • Heat shock proteins of interest include, without limitation, glycoprotein 96 (gp96), heat shock protein (hsp) 90, hsp70, hspl lO, glucose-regulated protein 170 (grpl 70) and calreticulin.
  • Immunogenic stimuli can include one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, more) heat shock proteins isolated from tumor cells or infected cells.
  • Such tumor or infected cells are preferably, but not necessarily, from the same subject (i) whose T cell responsiveness is to be enhanced by a method of the invention or (ii) from whom T cells (whose responsiveness is to be enhanced by a method of the invention) were obtained.
  • the tumor or infected cells can also be obtained, for example, from another individual having the same or a related tumor- type or infection as the subject.
  • the heat shock protein can be isolated from mammalian cells expressing a transcriptosome prepared from tumor cells or infected cells of interest.
  • Immunogenic molecules can be derived from a wide range of infectious microorganisms. Examples of relevant microorganisms include any of those listed above.
  • Relevant microbial proteins include, without limitation, the B subunit of heat labile enterotoxin of E. coli [Konieczny et al. (2000) FEMS Immunol. Med. Microbiol. 27(4):321-332], heat-shock proteins, e.g., the Y. enterocoliiica heat shock protein 60 [Konieczny et al. (2000) supra; Mertz et al. (2000) J. Immunol. 164(3): 1529-1537] and M.
  • tuberculosis heat-shock proteins hsp60 and hsp70 the Chlamydia trachomatis outer membrane protein [Ortiz et al. (2000) Infect. Immun. 68(3): 1719-1723], the B. burgdorferi outer surface protein [Chen et al. (1999) Arthritis Rheum. 42(9):1813-1823], the L. major GP63 [White et al. (1999) Vaccine 17(17):2150-2161 (and published erratum in Vaccine 17(20-21):2755)], the N. meningitidis meningococcal serotype 15 PorB protein [Delvig et al. (1997) Gin. Immunol.
  • CTL are by virtue of their ability to kill target cells infected with any of a wide variety of intracellular pathogens (e.g., vimses, or intracellular bacteria and protozoans) potent mediators of immunity to such pathogens.
  • pathogens e.g., vimses, or intracellular bacteria and protozoans
  • helper T cells release a wide variety of cytokines that mediate pathogen-destructive inflammatory reponses.
  • immunogenic stimuli useful in the invention can be any of a wide variety of tumor cells, APC “primed” with tumor cells, hybrid cells (see above), tumor-associated antigens (TAA), peptide-epitopes of such TAA, and APC “primed” with TAA or peptide-epitopes of them.
  • TAA tumor-associated antigens
  • a "TAA” is a molecule (e.g., a protein molecule) that is expressed by a tumor cell and either (a) differs qualitatively from its counterpart expressed in non ⁇ al cells, or (b) is expressed at a higher level in tumor cells than in normal cells.
  • a TAA can differ (e.g., by one or more amino acid residues where the molecule is a protein) from, or it can be identical to, its counterpart expressed in normal cells. It is preferably not expressed by normal cells. Alternatively, it is expressed at a level at least two-fold higher (e.g., a two-fold, three-fold, five-fold, ten- fold, 20-fold, 40-fold, 100-fold, 500-fold, 1, 000- fold, 5,000-fold, or 15,000-fold higher) in a tumor cell than in the tumor cell's normal counterpart.
  • two-fold higher e.g., a two-fold, three-fold, five-fold, ten- fold, 20-fold, 40-fold, 100-fold, 500-fold, 1, 000- fold, 5,000-fold, or 15,000-fold higher
  • tumors examples include, without limitation, hematological cancers such as leukemias and lymphomas, neurological tumors such as astrocytomas or glioblastomas, melanoma, breast cancer, lung cancer, head and neck cancer, gastrointestinal tumors such as gastric or colon cancer, liver cancer, renal cell cancer, pancreatic cancer, genitourinary tumors such ovarian cancer, vaginal cancer, bladder cancer, testicular cancer, prostate cancer or penile cancer, bone tumors, and vascular tumors.
  • hematological cancers such as leukemias and lymphomas
  • neurological tumors such as astrocytomas or glioblastomas, melanoma
  • breast cancer such as astrocytomas or glioblastomas, melanoma
  • lung cancer head and neck cancer
  • gastrointestinal tumors such as gastric or colon cancer
  • liver cancer liver cancer
  • renal cell cancer pancreatic cancer
  • genitourinary tumors such as ovarian cancer, vaginal cancer, bladder
  • TAA include, without limitation, carcinoembryonic antigen (CEA), prostate specific antigen (PSA), MAGE (melanoma antigen) 1-4, 6 and 12, MUC (mucin) (e.g., MUC-1, MUC-2, etc.), tyrosinase, MART (melanoma antigen), Pmel 17(gpl00), GnT-V intron V sequence (N-acetylglucoaminyltransferase V intron V sequence), Prostate Ca psm, PRAME (melanoma antigen), ⁇ -catenin, MUM-l-B (melanoma ubiquitous mutated gene product), GAGE (melanoma antigen) 1, BAGE (melanoma antigen) 2-10, C-ERB2 (Her2/neu), EBNA (Epstein-Barr Virus nuclear antigen) 1-6, gp75, human papilloma vims (HPV) E6 and E7, p53,
  • fusion proteins containing, as one domain, all or a portion of any of the polypeptide enhancing compounds (e.g., B7-H1), immunogenic stimuli, or non-specifically acting factors (e.g. cytokines) listed above. Additional domains in such fusion proteins can be additional functional domains or signal peptides. Such fusion proteins can also contain immunoglobulin heavy chain constant regions (e.g., mouse IgG2a or human IgGl heavy chain constant regions) or portions of such constant regions (e.g., CH2 and/or CH3 domains). Useful additional domains include those that facilitate purification of the peptide, e.g., a hexahistidine sequence.
  • Polypeptide enhancing compounds, immunogenic stimuli, and non- specifically acting factors useful for the invention include those described above, but modified for in vivo use by the addition, at the amino- and/or carboxyl -terminal ends, of a blocking agent to facilitate survival of the relevant polypeptide in vivo. This can be useful in those situations in which the peptide termini tend to be degraded by proteases prior to cellular uptake.
  • blocking agents can include, without limitation, additional related or unrelated peptide sequences that can be attached to the amino and/or carboxyl terminal residues of the peptide to be administered. This can be done either chemically during the synthesis of the peptide or by recombinant DNA technology by methods familiar to artisans of average skill.
  • blocking agents such as pyroglutamic acid or other molecules known in the art can be attached to the amino and/or carboxyl terminal residues, or the amino group at the amino terminus or carboxyl group at the carboxyl tenninus can be replaced with a different moiety.
  • the peptide compounds can be covalently or noncovalently coupled to pharmaceutically acceptable "carrier" proteins prior to administration.
  • Peptidomimetic compounds that are designed based upon the amino acid sequences of polypeptides of interest.
  • Peptidomimetic compounds are synthetic compounds having a three-dimensional conformation (i.e., a "peptide motif) that is substantially the same as the three-dimensional conformation of a selected peptide.
  • the peptide motif provides the peptidomimetic compound with the ability to activate an immune response (in the case of immunogenic stimuli) and enhance an immune response (in the case of the enhancing compounds).
  • Peptidomimetic compounds can have additional characteristics that enhance their in vivo utility, such as increased cell permeability and prolonged biological half-life.
  • the peptidomimetics typically have a backbone that is partially or completely non-peptide, but with side groups that are identical to the side groups of the amino acid residues that occur in the peptide on which the peptidomimetic is based.
  • Several types of chemical bonds e.g., ester, thioester, thioamide, retroamide, reduced carbonyl, dimethylene and ketomethylene bonds, are known in the art to be generally useful substitutes for peptide bonds in the construction of protease-resistant peptidomimetics.
  • Molecules useful as enhancing compounds, immunogenic stimuli, and non- specifically acting factors can be produced by any of a wide range of methods known in the art. They can be purified from natural sources (e.g., from any of the cancer cells or infectious microorganisms listed herein). Smaller peptides (fewer than 100 amino acids long) and other non-protein molecules can be conveniently synthesized by standard chemical means known to those in the art. In addition, both polypeptides and peptides can be manufactured by standard in vitro recombinant DNA techniques and in vivo transgenesis using nucleotide sequences encoding the appropriate polypeptides or peptides (see Nucleic Acids section below).
  • constmct expression vectors containing relevant coding sequences and appropriate transcriptional/translational regulatory elements. See, for example, the techniques described in Sambrook et al., Molecular Cloning: A Laboratory Manual (2nd Ed.) [Cold Spring Harbor Laboratory, N.Y., 1989], and Ausubel et al., Current Protocols in Molecular Biology [Green Publishing Associates and Wiley Interscience, N.Y., 1989].
  • the transcriptional/translational regulatory elements referred to above include but are not limited to inducible and non-inducible promoters, enhancers, operators and other elements that are known to those skilled in the art and that drive or otherwise regulate gene expression.
  • Such regulatory elements include but are not limited to the cytomegalovirus hCMV immediate early gene, the early or late promoters of SV40 adenovirus, the lac system, the trp system, the TAC system, the TRC system, the major operator and promoter regions of phage A, the control regions of fd coat protein, the promoter for 3-phosphoglycerate kinase, the promoters of acid phosphatase, and the promoters of the yeast ⁇ -mating factors.
  • the expression systems that may be used for purposes of the invention include but are not limited to microorganisms such as bacteria (for example, E. coli and B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA, or cosmid DNA expression vectors containing nucleic acid molecules encoding enhancing agents or immunogenic stimuli; yeast (for example, Saccharomyces and Pichia) transformed with recombinant yeast expression vectors containing a nucleic acid encoding enhancing agents or immunogenic stimuli; insect cell systems infected with recombinant virus expression vectors (for example, baculovirus) containing a nucleic acid encoding enhancing agents or immunogenic stimuli; plant cell systems infected with recombinant virus expression vectors (for example, cauliflower mosaic virus (CaMV) or tobacco mosaic vims (TMV)) or transfonued with recombinant plasmid expression vectors (for example, Ti plasmid) containing a nucle
  • Cells transfected or transduced with the expression vectors of the invention can then be used, for example, for large or small scale in vitro manufacture of enhancing agents or immunogenic stimuli by methods known in the art. In essence, such methods involve culturing the cells under conditions that maximize production of the polypeptide and isolating the polypeptide from the culture, i.e., the cells and/or the culture medium.
  • antibodies are known in the art. For example, they can be produced by immunizing animals with a substance of interest (e.g., B7-H1).
  • a useful antibody can be a polyclonal antibody present in the serum or plasma of an animal (e.g., human, non-human primate, mouse, rabbit, rat, guinea pig, sheep, horse, goat, cow, pig, or bird) which has been injected with the substance of interest, and optionally an adjuvant.
  • Polyclonal antibodies can be isolated from serum or plasma by methods known in the art.
  • Monoclonal antibodies (mAb) can also be produced. Methods of making and screening monoclonal antibodies are well known in the art.
  • the resultant antibody can be produced by a number of methods known in the art.
  • the hybridoma can be cultured in vitro in a suitable medium for a suitable length of time, followed by the recovery of the desired antibody from the supernatant.
  • the length of time and medium are known or can be readily determined.
  • Polyclonal and monoclonal antibodies can be manufactured in large amounts by methods known in the art.
  • large animals e.g., sheep, pigs, goats, horses, or cows
  • Serum can be isolated from the blood of animals producing an antibody with the appropriate activity.
  • polyclonal antibodies can be purified from such sera by methods known in the art.
  • Monoclonal antibodies can also be produced in large amounts in vitro using, for example, bioreactors or in vivo by injecting appropriate animals with the relevant hybridoma cells.
  • mice or rats can be injected intraperitoneally (i.p.) with the hybridoma cells and, after a time sufficient to allow substantial growth of the hybridoma cells and secretion of the monoclonal antibody into the blood of the animals, they can be bled and the blood used as a source of the monoclonal antibody.
  • mice or rats can be injected intraperitoneally (i.p.) with the hybridoma cells and, after a time sufficient to allow substantial growth of the hybridoma cells and secretion of the monoclonal antibody into the blood of the animals, they can be bled and the blood used as a source of the monoclonal antibody.
  • an inflammatory substance such as pristane
  • peritoneal exudates containing the monoclonal antibodies can develop in the animals. The peritoneal exudates can then be "tapped" from the animals and used as a source of the appropriate monoclonal antibody.
  • the enhancing compounds, immunogenic stimuli, and non-specifically acting factors be purified.
  • Methods for purifying biological macromolecules are known in the art.
  • the degree of purity of the macromolecules can be measured by any appropriate method, e.g., column chromatography, polyacrylamide gel electrophoresis, or HPLC analysis.
  • the methods of the invention can be performed in vitro, in vivo, or ex vivo. In vitro application of the methods can be useful, for example, in basic scientific studies of apoptosis. They can also be useful as assays for T cell responsiveness to a tumor- specific or infectious microorganism-derived antigen where T cell proliferation can be low and possibly undetectable in the absence of a compounds that contain agents that interfere with the physical interaction between a B7-H1 and a T cell.
  • the methods of the invention can be used as diagnostic assays for cancers and infectious disease.
  • the methods can also be useful for growing up large numbers of T cells (e.g., tumor infiltrating lymphocytes (TIL)) for adoptive immunotherapy of cancer or infectious diseases.
  • TIL tumor infiltrating lymphocytes
  • lymphoid cells consisting of or including T cells obtained from a mammalian subject are cultured with any of the above described enhancing compounds and immunogenic stimuli.
  • the lymphoid cells can be from a subject pre-exposed to a relevant antigen (in any of the fomis described above); alternatively, the donor of the lymphoid cells need not have been exposed to the antigen.
  • the cultures can also be supplemented with one or more cytokines or growth factors listed herein.
  • the cultures can also be monitored at various times to ascertain whether the desired level of immune reactivity (e.g., CTL or helper T cell activity) has been attained.
  • the methods of the invention will preferably be in vivo or ex vivo (see below).
  • the methods are generally useful in enhancing T cell responsiveness in subjects with cancer and infectious diseases.
  • the enhancement of T cell responsiveness effected by the methods of the invention can result in decreased cancer cell or infectious microorganism proliferation.
  • the methods of the invention can be applied to mammalian subjects (e.g., human breast cancer or melanoma patients) alone or in conjunction with other drugs and/or radiotherapy.
  • the methods of the invention can be applied to a wide range of species, e.g., humans, non-human primates (e.g., monkeys or chimpanzees), horses, cattle, pigs, sheep, goats, dogs, cats, rabbits, guinea pigs, hamsters, rats, and mice.
  • non-human primates e.g., monkeys or chimpanzees
  • horses cattle, pigs, sheep, goats, dogs, cats, rabbits, guinea pigs, hamsters, rats, and mice.
  • methods of invention enhance T cell responsiveness, they are generally useful for enhancing immune responses.
  • Such immune responses can be prophylactic or therapeutic.
  • the responses enhanced need have neither prophylactic nor therapeutic efficacy. They can be used, for example, (a) to produce large numbers of activated T cells for use in basic scientific studies of T cell activity; or (b) to enhance T cell responses that provide helper activity for antibody-producing B cells and thereby facilitate the production of large quantities of antibodies in mammals (e.g., rabbits, goats, sheep, or horses) that are subsequently isolated from the animals and used for purposes such as antigen detection or purification, or (c) for immunization of animals (e.g., mice, rats, or hamsters) with a view to making monoclonal antibodies.
  • mammals e.g., rabbits, goats, sheep, or horses
  • the methods of the invention can be used, for example, for prophylaxis or therapy against (a) infectious diseases due to any of the infectious agents listed herein; or (b) cancers such as any of those listed herein.
  • infectious diseases due to any of the infectious agents listed herein; or (b) cancers such as any of those listed herein.
  • appropriate methods can be used for prophylaxis, h regard to infectious microorganisms, the methods can be particularly useful in the prevention and/or therapy of diseases involving intracellular microorganisms (i.e.
  • infectious agents that replicate inside a cell e.g., viruses such as influenza vims or HIV, intracellular bacteria such M. tuberculosis, and intracellular protozoans such as P. falciparum or any of the other infectious agents listed herein.
  • viruses such as influenza vims or HIV
  • intracellular bacteria such M. tuberculosis
  • intracellular protozoans such as P. falciparum or any of the other infectious agents listed herein.
  • prophylaxis can mean complete prevention of the symptoms of a disease, a delay in onset of the symptoms of a disease, or a lessening in the severity of subsequently developed disease symptoms. "Prevention” should mean that symptoms of the disease (e.g., an infection) are essentially absent.
  • therapy can mean a complete abolishment of the symptoms of a disease or a decrease in the severity of the symptoms of the disease.
  • a "protective" immune response is an immune response that is prophylactic and/or therapeutic. In one in vivo approach, the enhancing compound itself, and optionally an immunogenic stimulus and/or one or more nonspecifically acting factors (see above), is administered to the subject.
  • the substances to be administered will be suspended in a pharmaceutically-acceptable carrier (e.g., physiological saline) and administered orally or by intravenous infusion, or injected subcutaneously, intramuscularly, intrathecally, intraperitoneally, intrarectally, intravaginally, intranasally, intragastrically, intratracheally, or intrapulmonarily.
  • a pharmaceutically-acceptable carrier e.g., physiological saline
  • intravenous infusion e.g., physiological saline
  • the dosage required depends on the choice of the route of administration; the nature of the formulation; the nature of the patient's illness; the subject's size, weight, surface area, age, and sex; other drugs being administered; and the judgment of the attending physician. Suitable dosages are in the range of 0.01-1,000.0 ⁇ g/kg. Wide variations in the needed dosage are to be expected in view of the variety of enhancing compounds available and the differing efficiencies of various routes of administration. For example, oral administration would be expected to require higher dosages than administration by intravenous, injection. Variations in these dosage levels can be adjusted using standard empirical routines for optimization as is well understood in the art.
  • Administrations can be single or multiple (e.g., 2-, 3-, 4-, 6-, 8-, 10-, 20-, 50-,100-, 150-, or more fold).
  • Encapsulation of the substances in a suitable delivery vehicle e.g., polymeric microparticles or implantable devices
  • a suitable delivery vehicle e.g., polymeric microparticles or implantable devices
  • hnmunogenic stimuli, and/or non-specifically acting factors can be administered before at the same time as, or after administration of the enhancing compounds.
  • adjuvants can be used together with the immunogenic stimuli.
  • Suitable adjuvants include cholera toxin (CT), E. coli heat labile toxin (LT), mutant CT (MCT) [Yamamoto et al. (1997) J. Exp. Med. 185:1203-1210] and mutant E. coli heat labile toxin (MLT) (Di'Tommaso et al. (1996) Infect. Immunity 64:974-979].
  • CT cholera toxin
  • LT E. coli heat labile toxin
  • MCT mutant CT
  • MLT mutant E. coli heat labile toxin
  • MLT mutant E. coli heat labile toxin
  • one or more of the above-listed cytokines or growth factors can be administered (by any of the routes recited herein) to the subject, before, at the same time as, or after administration of the enhancing agents and, optionally, immunogenic stimuli.
  • tumor cells, APC, or hybrid cells can express on their surface or secrete either (a) one or more recombinant costimulatory molecules (e.g., B7.1, B7.2, B7-H2, B7-H3, or B7-H4) and/or (b) one or more recombinant cytokines or recombinant growth factors such as those listed above, e.g., GM-CSF.
  • Cells expressing on their surface or secreting the above recombinant molecules will have been transfected (stably or transiently) or transfonued with one or more nucleic acids (e.g., expression vectors) encoding the molecules.
  • a polynucleotide containing a nucleic acid sequence encoding a polypeptide enhancing compound can be delivered to cancer cells or a site of infection in a mammal.
  • Expression of the coding sequence will preferably be directed to lymphoid tissue of the subject by, for example, delivery of the polynucleotide to the lymphoid tissue.
  • Expression of the coding sequence can be directed to any cell in the body of the subject. However, expression will preferably be directed to cells in the vicinity of the tumor cells whose responsiveness it is desired to inhibit.
  • expression of the coding sequence can be directed to the tumor cells themselves. This can be achieved by, for example, the use of polymeric, biodegradable microparticle or microcapsule delivery devices known in the art.
  • liposomes prepared by standard methods.
  • the vectors can be incorporated alone into these delivery vehicles or co-incorporated with tissue-specific or tumor-specific antibodies.
  • tissue-specific or tumor-specific antibodies can be prepared by electrostatic or covalent forces.
  • Poly-L-lysine binds to a ligand that can bind to a receptor on target cells [Cristiano et al. ( 1995), J. Mol. Med. 73, 479].
  • tissue specific targeting can be achieved by the use of tissue-specific transcriptional regulatory elements (TRE) which are known in the art. Delivery of "naked DNA" (i.e., without a delivery vehicle) to an intramuscular, intradermal, or subcutaneous site is another means to achieve in vivo expression.
  • TRE tissue-specific transcriptional regulatory elements
  • the nucleic acid sequence encoding the enhancing compound polypeptide of interest with an initiator methionine and optionally a targeting sequence is operatively linked to a promoter or enhancer-promoter combination.
  • Short amino acid sequences can act as signals to direct proteins to specific intracellular compartments. Such signal sequences are described in detail in U.S. Patent No. 5,827,516, incorporated herein by reference in its entirety. Enhancers provide expression specificity in terms of time, location, and level.
  • an enhancer can function when located at variable distances from the transcription initiation site, provided a promoter is present.
  • An enhancer can also be located downstream of the transcription initiation site. To bring a coding sequence under the control of a promoter, it is necessary to position the translation initiation site of the translational reading frame of the peptide or polypeptide between one and about fifty tiucleotides downstream (3') of the promoter.
  • the coding sequence of the expression vector is operatively linked to a transcription terminating region.
  • Suitable expression vectors include plasmids and viral vectors such as herpes vimses, retrovimses, vaccinia viruses, attenuated vaccinia vimses, canary pox viruses, adenovimses and adeno-associated vimses, among others.
  • Polynucleotides can be administered in a phannaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are biologically compatible vehicles that are suitable for administration to a human, e.g., physiological saline or liposomes.
  • a therapeutically effective amount is an amount of the polynucleotide that is capable of producing a medically desirable result (e.g., decreased proliferation of cancer cells) in a treated animal.
  • the dosage for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently.
  • a preferred dosage for administration of polynucleotide is from approximately 10 6 to 10 12 copies of the polynucleotide molecule. This dose can be repeatedly administered, as needed. Routes of administration can be any of those listed above.
  • Lymphoid cells e.g., peripheral blood mononuclear cells (PBMC)
  • PBMC peripheral blood mononuclear cells
  • the cultures can also contain any of a variety of cytokines (see above)
  • the lymphoid cells containing highly activated T cells are then introduced into the same or a different patient.
  • Another ex vivo strategy can involve transfecting or transducing cells obtained from the subject with a polynucleotide encoding an enhancing compound.
  • the transfected or transduced cells are then returned to the subject.
  • the cells can be any of a wide range of types including, without limitation, hemopoietic cells (e.g., bone marrow cells, macrophages, monocytes, dendritic cells, T cells, or B cells), fibroblasts, epithelial cells, endothelial cells, keratinocytes, or muscle cells.
  • hemopoietic cells e.g., bone marrow cells, macrophages, monocytes, dendritic cells, T cells, or B cells
  • fibroblasts e.g., epithelial cells, endothelial cells, keratinocytes, or muscle cells.
  • Such cells act as a source of the enhancing compound for as long as they survive in the subject.
  • tumor cells preferably obtained from the subject but potentially from an individual other than the subject, can be transfected or transformed by a vector encoding an enhancing compound.
  • the tumor cells preferably treated with an agent (e.g., ionizing irradiation) that ablates their proliferative capacity, are then introduced into the patient, where they secrete the enhancing compound.
  • an agent e.g., ionizing irradiation
  • ex vivo methods include the steps of harvesting cells from a subject, culturing the cells, transducing them with an expression vector, and maintaining the cells under conditions suitable for expression of the enhancing compound. These methods are known in the art of molecular biology.
  • the transduction step is accomplished by any standard means used for ex vivo gene therapy, including calcium phosphate, lipofection, electroporation, viral infection, and biolistic gene transfer. Alternatively, liposomes or polymeric microparticles can be used.
  • Cells that have been successfully transduced can then be selected, for example, for expression of the coding sequence or of a drug resistance gene. The cells may then be lethally irradiated (if desired) and injected or implanted into the subject.
  • an immunogenic stimulus can be provided in the fonn of a peptide-epitope and the enhancing compound in the form of either a nucleic acid encoding it or cells transformed with a nucleic acid encoding it.
  • the methods of the invention can be applied to any of the diseases and species listed here. Methods to test whether a given modality is therapeutic for or prophylactic against a particular disease are known in the art. Where a therapeutic effect is being tested, a test population displaying symptoms of the disease (e.g., cancer patients) is treated by a method of the invention, using any of the above described strategies. A control population, also displaying symptoms of the disease, is treated, using the same methodology, with a placebo. Disappearance or a decrease of the disease symptoms in the test subjects would indicate that the method was therapeutic.
  • a test population displaying symptoms of the disease e.g., cancer patients
  • a control population also displaying symptoms of the disease, is treated, using the same methodology, with a placebo. Disappearance or a decrease of the disease symptoms in the test subjects would indicate that the method was therapeutic.
  • the method can be tested for prophylactic efficacy. In this situation, prevention of onset of disease symptoms is tested.
  • Analogous strategies can be used to test for the efficacy of the methods in the prophylaxis of a wide variety of infectious diseases, e.g., those involving any of the microorganisms listed above.
  • mAbs were purified from hybridoma culture supernatants by adsorption to and elution from Protein G -Sepharose (Pierce, Rockford, 1L), and were dialyzed in LPS-free PBS. Clones 2H1 and 5H1 were used for the studies described in the following examples. Both mAbs are IgGl and have similar staining patterns and blocking activity.
  • the PD-lIg fusion protein was prepared as described [Chapoval et al. (2000) Methods Mol. Med. 45:247-255].
  • F(ab') 2 fragments of anti- hB7-Hl mAb (5H1) were prepared by digestion and purification using an Immobilized Pepsin Kit (Pierce, Rockford, IL).
  • Anti-2C mAb specific for the T cell receptor (TCR) expressed on T cells of 2C TCR transgenic mice was purified from culture supernatants of hybridoma 1B2 [Sykulev et al. (1994) Proc. Natl. Acad. Sci. USA 91: 1 1487-11491] and conjugated with phycoerythrin (PE).
  • the mAbs specific for CD8 (RPA-T8), Fas (DX2) and Fas ligand (NOK-1) were purchased from BD PharMingen (San Diego, CA) and the rabbit anti-human TRAIL polyclonal antibody was purchased from Alexis Biochemicals (San Diego, CA).
  • the phenotypes of T cells and tumor cells were analyzed by staining with specific mAb and fluorescence was analyzed by FFC using a FACScan (Becton Dickinson, Mountain View, CA) and Cell Quest software (Becton Dickinson, San Jose, CA).
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • RosetteSep Human T cell enrichment cocktail >95% purity
  • T cell proliferation was determined after adding 1.0 mCi H-thymidine ( H-TdR) at least 18 firs before cell harvest. Incorporation of 3 H-TdR was measured with a MicroBeta TriLux liquid scintillation counter (Wallac, Finland).
  • Tumor-specific CD4+ and CD8+ T cell clones were generated by in vitro stimulation with dendritic cells loaded with corresponding tumour antigens or peptide epitopes [Kobayashi et al. (2000) Cancer Res. Vol. 60: 5228-5236; Lu et al. (2000) Cancer Res. Vol. 60: 5223-5227].
  • Ml 5 is a human CD8+ CTL clone that specifically recognizes an HL A- A2 -restricted epitope (IMDQVPFSV) (SEQ ID NO: 1 ) of the gplOO melanoma antigen.
  • M99 is an HLA-B7-restricted CD8+ CTL clone [Lu et al. (2000) Cancer Res. 60:5223-5227] that recognizes an epitope (IPQQHTQVL) (SEQ ID NO:2) of carcinoembryonic antigen.
  • IPQQHTQVL epitope
  • T cells 4 x 10 7 CD8 + T cells were purified from spleen and lymph nodes of 2C mice by MACS beads (Milteni Biotec, Milteni, Germany) and subsequently stimulated with irradiated BALB/c spleen cells in the presence of 10 U/ml human IL-2. Three days later, T cells were harvested, depleted of dead cells by centrifugation over Lympholyte-M (Cedarlane Laboratories, Hornby, Ontario, Canada), and used for adoptive transfer.
  • MACS beads Milteni Biotec, Milteni, Germany
  • the human CD4+ T cell clones 4D16 and 8G9, and CD8+ T cell line (M15) were cultured for 2 weeks as described previously [Kobayashi et al. (2000) Cancer
  • IFN-E treated SKmel-28 were pulsed with 10 mg/ml of MAGE-3 pl46-160 peptide for 2 h at 37°C. After washing, SKmel-28 cells were incubated with the control mlgGl or anti-B7-Hl mAb (2H1) for 30 min at 37°C before co-culture with T cells.
  • Pre-activated CD4+ T cells at 2 x 105 cells/well were co-cultured at 37°C for 6 days with tumor cells at 2 x 104 cells/well (T cell to tumor cell ratio of 10:1) in the presence of blocking agents or control mouse IgG 1.
  • the CTL (Ml 5 or M99 clones) (2 x 10 5 cells/well) were cultured with irradiated tumor cells (1 -5 x 10 4 cells/well) for 4-5 days. Monoclonal antibodies, fusion protein and control IgG were included in the culture ab initio. Cells were harvested at the indicated times and stained with Annexin V and anti-CD8 mAb. The cells were analyzed by a FACScan and apoptosis was calculated as the percentage of Annexin V-staining cells gated in the CD8-expressing populations.
  • human peripheral blood T cells (2 x 105 cells/well) were cultured with irradiated NCI-H292 cells (4 xl04 cells/well) for 5 days.
  • Blocking agents such as anti-l ⁇ B7-Hl mAb (2H1), l ⁇ B7-HHg, PD-lIg fusion proteins or mouse IgGl (as control) were added (20 mg/ml) at the begim ing of cell co-culture.
  • Harvested cells were stained with 7-AAD, anti-CD4 and anti-CD8 mAbs. Both early and late apoptotic cells were gated as R2 and viable cells as R3.
  • the cells in the R2 and R3 gates were analyzed for expression of CD4 or CD8.
  • human peripheral blood T cells were isolated and purified by RosetteSep Human T cell enrichment cocktail (Stemcell, Canada) to obtain purified T cells (>95%).
  • T cells at a concentration of 2 x 10 5 /well (3 wells per group) were cultured in 96- well flat-bottomed tissue culture plates that had been pre-coated overnight with 0.5 ⁇ g/ml of anti-CD3 mAb (clone HIT3a, PharMingen, Palo Alto, CA) in the presence of immobilized hB7-HHg or control Ig (mouse IgG2a) for 72 hours. Both the hB7-HHg and control Ig were pre- coated onto the plates at a concentration of 10 ⁇ g/ml.
  • human PD-lIg (30 ⁇ g/ml), a neutralizing mAb specific for human IL-10 ( 5 ⁇ g/ml; JES3-9D7; PharMingen), or a neutralizing mAb specific for human Fas ligand ( 8 ⁇ g /ml, NOK-1, PharMingen) were added at the beginning of culture.
  • An immobilized fusion protein comprising the extracellular portion of hB7-Hl and the constant region of the heavy chain of human IgGl (hB7-Hl Ig) costimulated the growth of freshly isolated CD3 + T cells from human peripheral blood mononuclear cells (PBMC) activated with a suboptimal TCR stimulus, i.e., with a low concentration of antibody specific for the CD3 molecule. Under such conditions, increased 3 H-thymidine incorporation and the number of recovered viable cells indicated that the T cells proliferated for up to 64 hrs (Figs. 1 A and B).
  • PBMC peripheral blood mononuclear cells
  • Monoclonal antibodies specific for hB7-Hl were used to determine whether fiB7-Hl protein expression parallels of mRNA expression as previously determined. No l ⁇ B7-Hl protein expression was detected in any normal solid tissue examined including breast, colon, pancreas, kidney, uterus, skeletal muscle, lung, liver, tonsil, and intestine. Interestingly, l B7-Hl -expressing macrophages were observed in liver, lung and tonsil.
  • hB7-Hl mRNA in these tumor cell lines was confirmed by the reverse transcriptase- coupled polymerase chain reaction (RT-PCR) using hB7-Hl-specific primers (data not shown). Table 1. Expression of hB7-Hl in human tumor cell lines
  • hB7-Hl expression demonstrated hB7-Hl expression in a majority of freshly isolated human lung carcinomas (20/21 patients), ovarian carcinomas (20/23 patients), colon carcinomas (10/19 patients) and melanomas (22/22 patients) (summarized in Table 2).
  • B7-H1 expression was observed in the plasma membrane, cytoplasm or both.
  • hB7-Hl expression was usually focal with adjacent normal tissues negative.
  • hB7-Hl was evident on metastatic melanoma cells residing in lymph nodes, but not on adjacent lymphocytes.
  • T cells were purified from peripheral blood mononuclear cells of healthy donors and incubated for 5 days with irradiated cells of the NCI-H292 line, a hB7-Hl + FasL " mucoepidermoid lung carcinoma cell line (Fig. 2A). T cells undergoing apoptosis were detected by triple staining with 7-amino- actinomycin D (7-AAD) and CD4 and CD8-specific mAbs to identify the subsets of apoptotic T cells.
  • 7-AAD 7-amino- actinomycin D
  • Fig. 2B shows that 22.4 % of T cells were apoptotic (7-AAD + ) in cultures containing NCI-H292 cells, while only 12.1 % of T cells were apoptotic when cultured with medium only. This difference represented an increase of 85 % in apoptotic T cells over the basal level.
  • CD4 + T cells accounted for >95% of apoptotic T cells while the viable population contained both CD4 + and CD8 + cells (Fig. 2A).
  • hB7-Hl on tumor cells promotes apoptosis of T cells
  • cells of the hB7-Hl -non-expressing human melanoma line, 624mel were transfected with a plasmid encoding full-length hB7-Hl cDNA.
  • hB7-Hl expression on transfected cells was confirmed by staining with anti-hB7-H 1 mAb (Fig. 3D).
  • purified human T cells had been co-cultured for 6 days with 624mel cells, there were significantly more apoptotic cells than when the same T cells were cultured under the same conditions with mock-transfected 624mel cells (data not shown).
  • tumor-associated hB7-Hl promotes apoptosis of T cells.
  • the increased apoptosis in T cells after prolonged exposure to tumor- associated l ⁇ B7- Hl in the above experiments could have been due to allogeneic antigen rather than tumor antigen recognition by T cells.
  • T cells specific for tumor antigens respond similarly to tumor-associated hB7-Hl, induction of apoptosis in the 8G9 CD4 + human T cell clone, which recognizes an HLA-DR4-restricted helper T cell epitope of tumour antigen MAGE-3
  • the MAGE-3 + human melanoma line, SKmel-28 expresses a low level of HLA-DR4 that is upregulated by treatment with IFN- ⁇ .
  • IFN- ⁇ treatment of SKmel-28 also upregulated hB7-Hl and HLA-DQ but not Fas ligand or TRAIL (Fig. 3 A).
  • IFN- ⁇ treated SKmel- 28 cells stimulates cells of the 8G9 T cell clone to release GM-CSF (data not shown). Incubation of 8G9 T cell clone with IFN- ⁇ treated Skmel-28 cells for 6 days led to a reduced number of viable cells and an increase in apoptotic T cells (50-55%).
  • Hl/624mel cells were again used.
  • Parental, untransfected 624mel cells express the gplOO tumor antigen and MHC class 1 molecules (HLA-A2), [Rivoltini et al. (1995), Cancer Res. 55:3149-3157] but not hB7-Hl, even after treatment with IFN-D (Fig. 3D and Table 1).
  • 624mel cells do not express Fas ligand (FasL) or TRAIL proteins, which are known to participate in the induction of T cell apoptosis [Nagata et al. (1995) Science 267: 1449-1456; Jeremias et al. (1998) J. Tmmunol.
  • a human CD8 + cytolytic T cell (CTL) clone (M15) specific for a gplOO peptide epitope was generated from peripheral blood mononuclear cells obtained from a healthy HLA-A2-expressing donor by a previously described method involving repeated stimulation in vitro with the HLA-A2-restricted gplOO epitope peptide [Lu et al. (2000) Cancer Res. 60:5223- 5227]. This gplOO peptide-reactive clone specifically lyses gpl00+ tumor cells (Fig. 5C and unpublished data).
  • Fig. 4 A shows that when Ml 5 CTL were co-cultured with Mock/624mel cells for 5 days, 14 ⁇ 5.6% of the M15 CTL cells underwent apoptotic death. However, when co-cultured with B7-Hl/624mel cells, 23 ⁇ 4.7% of the Ml 5 CTL population underwent apoptotic death (Fig. 4A). This represented a relative increase of 62% (P ⁇ 0.05).
  • the ligand specificity of this phenomenon was demonstrated by including in the culture medium anti-l ⁇ B7-Hl mAb or F(ab') 2 fragments of the mAb.
  • the anti- hB7-Hl mAb inhibited apoptosis of T cells by >50% (P ⁇ 0.05) and increased the number of viable T cells 1.4-fold (P ⁇ .05).
  • Addition of PD-lIg fusion protein at high concentrations also significantly inhibited the induction of Ml 5 cell apoptosis by B7- Hl/624mel (Fig. 4C). Consistent with this observation, nearly all Mock/624mel cells were eliminated in 5-day cultures with Ml 5 CTL while B7-Hl/624mel target cells were unaffected. Inclusion of anti-hB7-Hl mAb in the cultures abrogated the resistance of B7-Hl/624mel cells to killing by T cells (Figs. 5A and B).
  • B7-Hl/624mel cells to M15-mediated lysis was compared in a 4-hour ⁇ l Cr release cytotoxicity assay.
  • Mock/624mel and B7-Hl/624mel were equally sensitive to the lysis by Ml 5 CTL.
  • there was no growth advantage of B7-Hl/624mel over Mock/624mel cells in a proliferation assay in vitro (data not shown).
  • HBL-100 a human breast cancer line
  • Fig. 6 A demonstrates that HBL- 100 cells express hB7-Hl , but do not express FasL or TRAIL on their surfaces (Fig. 6A).
  • HBL-100 cells express a CTL epitope
  • JPQQHTQVL (SEQ ID NO:2) that is derived from carcinoembryonic antigen (CEA), is HLA-B7-restricted, and is recognized and lysed by CD8+ CTL clone M99 [Lu et al. (2000) Cancer Res. 60:5223-5227].
  • B7-Hl /624mel cells the number of antigen-specific M99 CTL was reduced and apoptosis increased when they were incubated with HBL-100 cells. T cell death was inhibited when anti-B7-Hl mAb was included in the medium (Fig. 6B).
  • T cell apoptosis in vitro is a correlate of activated T cell deletion in vivo
  • a mouse model involving adoptive transfer of 2C T cell receptor (TCR) transgenic mouse T cells which recognize a p2Ca peptide in the context of L d MHC class I molecule on the P815 tumor line was used.
  • Mock-transfected P815 cells (Mock/P815) or P815 cells transfected with a vector that expressses mB7-Hl (B7-H1/P815) were injected intraperitoneally (i.p.) into immunodeficient RAG-1 " * mice to establish progressively growing tumors.
  • 2C T cells were activated by incubation in vitro with BALB/c (H-2 ) spleen cells in the presence of human IL-2 and then transferred i.p. into the P815-bearing mice. By 18 hours after initial contact with the tumor cells, the relative proportion of 2C T cells had greatly increased in mice harboring Mock/P815 cells and accounted for nearly 10% of total peritoneal cells.

Abstract

L'invention concerne des procédés permettant d'améliorer la capacité de réaction d'une cellule T. De tels procédés impliquent d'agir sur l'interaction entre une cellule T et une molécule B7-H1.
PCT/US2002/012733 2001-04-20 2002-04-22 Procedes d'amelioration de la capacite de reaction de cellules t WO2002086083A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002258941A AU2002258941A1 (en) 2001-04-20 2002-04-22 Methods of enhancing cell responsiveness

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US28513701P 2001-04-20 2001-04-20
US60/285,137 2001-04-20

Publications (2)

Publication Number Publication Date
WO2002086083A2 true WO2002086083A2 (fr) 2002-10-31
WO2002086083A3 WO2002086083A3 (fr) 2003-02-27

Family

ID=23092893

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/012733 WO2002086083A2 (fr) 2001-04-20 2002-04-22 Procedes d'amelioration de la capacite de reaction de cellules t

Country Status (3)

Country Link
US (2) US7794710B2 (fr)
AU (1) AU2002258941A1 (fr)
WO (1) WO2002086083A2 (fr)

Cited By (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007100098A1 (fr) 2006-03-03 2007-09-07 Kyoto University Multimere de domaine extracellulaire d'une molecule fonctionnelle de surface cellulaire
EP1895842A2 (fr) * 2005-06-17 2008-03-12 University of Rochester Procedes et compositions permettant d'ameliorer la memoire immune par blocage de l'effacement intrahepatique des lymphocytes t actives
WO2009089149A1 (fr) * 2008-01-03 2009-07-16 The Johns Hopkins University Antagonistes de b7-h1 (cd274) induisant l'apoptose de cellules tumorales
EP2206517A1 (fr) * 2002-07-03 2010-07-14 Ono Pharmaceutical CO., LTD. Compositions immunostimulantes contenant un anticorps anti-PD-L1
WO2010098788A2 (fr) * 2008-08-25 2010-09-02 Amplimmune, Inc. Antagonistes de pd-i et procédés de traitement d'une maladie infectieuse
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2011161699A2 (fr) 2010-06-25 2011-12-29 Aurigene Discovery Technologies Limited Composés modulateurs de l'immunosuppression
US20120201824A1 (en) * 2008-11-11 2012-08-09 Mariusz Wasik Compositions and Methods for Inhibiting an Oncogenic Protein to Enhance Immunogenicity
WO2012145493A1 (fr) 2011-04-20 2012-10-26 Amplimmune, Inc. Anticorps et autres molécules qui se lient à b7-h1 et à pd-1
WO2012168944A1 (fr) 2011-06-08 2012-12-13 Aurigene Discovery Technologies Limited Composés thérapeutiques pour une immunomodulation
WO2013056716A1 (fr) * 2011-10-17 2013-04-25 Herlev Hospital Immunothérapie par pd-l1
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
US8460927B2 (en) 1999-11-30 2013-06-11 Mayo Foundation For Medical Education And Research B7-H1 antibodies and method of use
WO2013132317A1 (fr) 2012-03-07 2013-09-12 Aurigene Discovery Technologies Limited Composés peptidomimétiques utilisés comme immunomodulateurs
WO2013144704A1 (fr) 2012-03-29 2013-10-03 Aurigene Discovery Technologies Limited Composés cycliques d'immunomodulation provenant de la boucle bc de pd1 humain
US8652465B2 (en) 2005-06-08 2014-02-18 Emory University Methods and compositions for the treatment of persistent infections
US8747833B2 (en) 2004-10-06 2014-06-10 Mayo Foundation For Medical Education And Research B7-H1 and methods of diagnosis, prognosis, and treatment of cancer
US8779105B2 (en) 2005-05-09 2014-07-15 Medarex, L.L.C. Monoclonal antibodies to programmed death 1 (PD-1)
WO2014194293A1 (fr) 2013-05-30 2014-12-04 Amplimmune, Inc. Méthodes améliorées de sélection de patients pouvant être soumis à des thérapies ciblant pd-1 ou b7-h4, et polythérapies associées
WO2015033299A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,2,4-oxadiazole utilisés comme immunomodulateurs
WO2015033301A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,3,4-oxadiazole et 1,3,4-thiadiazole servant d'immunomodulateurs
WO2015033303A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Composés peptidomimétiques cycliques utilisés comme immunomodulateurs
US9212224B2 (en) 2012-05-15 2015-12-15 Bristol-Myers Squibb Company Antibodies that bind PD-L1 and uses thereof
WO2016142833A1 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés 1,2,4-oxadiazoles et thiadiazoles utilisés comme immunomodulateurs
JP2017002077A (ja) * 2006-12-27 2017-01-05 エモリー ユニバーシティ 感染症および腫瘍を処置するための組成物および方法
WO2017009842A2 (fr) 2015-07-16 2017-01-19 Biokine Therapeutics Ltd. Compositions et méthodes pour le traitement du cancer
US9598491B2 (en) 2008-11-28 2017-03-21 Emory University Methods for the treatment of infections and tumors
US9683048B2 (en) 2014-01-24 2017-06-20 Novartis Ag Antibody molecules to PD-1 and uses thereof
WO2017106061A1 (fr) 2015-12-14 2017-06-22 Macrogenics, Inc. Molécules bispécifiques présentant une immunoréactivité par rapport à pd-1 et à ctla-4 et leurs procédés d'utilisation
WO2017106372A1 (fr) 2015-12-15 2017-06-22 Oncoimmune, Inc. Anticorps monoclonaux chimériques et humanisés anti-ctla4 humaine, et leurs utilisations
WO2017112741A1 (fr) 2015-12-22 2017-06-29 Novartis Ag Récepteur d'antigène chimérique (car) contre la mésothéline et anticorps contre l'inhibiteur de pd-l1 pour une utilisation combinée dans une thérapie anticancéreuse
WO2017203362A1 (fr) * 2016-05-25 2017-11-30 The Council Of The Queensland Institute Of Medical Research Inhibiteurs du point de contrôle immunitaire et lymphocytes t cytotoxiques pour le traitement du cancer
US9988452B2 (en) 2014-10-14 2018-06-05 Novartis Ag Antibody molecules to PD-L1 and uses thereof
WO2018194496A2 (fr) 2017-04-17 2018-10-25 Закрытое Акционерное Общество "Биокад" Anticorps monoclonal à pd-l1
US10160806B2 (en) 2014-06-26 2018-12-25 Macrogenics, Inc. Covalently bonded diabodies having immunoreactivity with PD-1 and LAG-3, and methods of use thereof
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
EP3456346A1 (fr) 2015-07-30 2019-03-20 MacroGenics, Inc. Molécules de liaison pd-1 et lag-3 et leurs procédés d'utilisation
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
EP3292873B1 (fr) 2013-02-22 2019-05-01 CureVac AG Combinaison de vaccination et d'inhibition de la voie pd-1
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
WO2019246110A1 (fr) 2018-06-20 2019-12-26 Incyte Corporation Anticorps anti-pd-1 et leurs utilisations
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
WO2020132560A2 (fr) 2018-12-21 2020-06-25 Aim Immunotech Inc. Compositions et méthodes pour cancérothérapie
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1
US11040948B2 (en) 2017-09-29 2021-06-22 Curis, Inc. Crystal forms of immunomodulators
WO2021138512A1 (fr) 2020-01-03 2021-07-08 Incyte Corporation Polythérapie comprenant des inhibiteurs d'a2a/a2b et de pd-1/pd-l1
US11078279B2 (en) 2015-06-12 2021-08-03 Macrogenics, Inc. Combination therapy for the treatment of cancer
WO2021158635A1 (fr) 2020-02-07 2021-08-12 Al Therapeutics, Inc. Compositions antivirales et procédés d'utilisation
US11136300B2 (en) 2017-10-11 2021-10-05 Aurigene Discovery Technologies Limited Crystalline forms of 3-substituted 1,2,4-oxadiazole
US11174315B2 (en) 2015-10-08 2021-11-16 Macrogenics, Inc. Combination therapy for the treatment of cancer
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
WO2022147092A1 (fr) 2020-12-29 2022-07-07 Incyte Corporation Polythérapie comprenant des inhibiteurs a2a/a2b, des inhibiteurs pd-1/pd-l1 et des anticorps anti-cd73
US11458195B2 (en) 2013-02-22 2022-10-04 Curevac Ag Combination of vaccination and inhibition of the PD-1 pathway
US11459394B2 (en) 2017-02-24 2022-10-04 Macrogenics, Inc. Bispecific binding molecules that are capable of binding CD137 and tumor antigens, and uses thereof
US11497734B2 (en) 2017-11-03 2022-11-15 Aurigene Discovery Technologies Limited Dual inhibitors of TIM-3 and PD-1 pathways
US11497735B2 (en) 2017-11-06 2022-11-15 Aurigene Discovery Technologies Limited Conjoint therapies for immunomodulation
US11673894B2 (en) 2018-02-27 2023-06-13 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as A2A / A2B inhibitors
US11840567B2 (en) 2017-10-03 2023-12-12 Joint Stock Company “Biocad” Bispecific antibodies with specific binding to CD47 and PD-L1
US11873304B2 (en) 2018-05-18 2024-01-16 Incyte Corporation Fused pyrimidine derivatives as A2A/A2B inhibitors
US11884665B2 (en) 2019-01-29 2024-01-30 Incyte Corporation Pyrazolopyridines and triazolopyridines as A2A / A2B inhibitors

Families Citing this family (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7030219B2 (en) 2000-04-28 2006-04-18 Johns Hopkins University B7-DC, Dendritic cell co-stimulatory molecules
US7794710B2 (en) * 2001-04-20 2010-09-14 Mayo Foundation For Medical Education And Research Methods of enhancing T cell responsiveness
CA2466279A1 (fr) * 2001-11-13 2003-05-22 Dana-Farber Cancer Institute, Inc. Agents modulant l'activite de cellules immunes et procedes d'utilisation associes
ES2369077T3 (es) * 2002-06-06 2011-11-25 Immunicum Ab Nuevo procedimiento y composición para producir una vacuna alogénica celular.
JP5714212B2 (ja) 2005-12-08 2015-05-07 メダレックス・リミテッド・ライアビリティ・カンパニーMedarex, L.L.C. O8eに対するヒトモノクローナル抗体
WO2007082144A2 (fr) * 2006-01-05 2007-07-19 Mayo Foundation For Medical Education And Research Methode permettant de detecter un cancer a l'aide de b7-h1 et de survivine
WO2007106571A2 (fr) * 2006-03-15 2007-09-20 Soper Bryan R Procedes de criblage et de cartographie de variations genotypiques et phenotypiques de cellules
WO2007124361A2 (fr) * 2006-04-20 2007-11-01 Mayo Foundation For Medical Education And Research B7-h1 soluble
AU2007342338A1 (en) * 2006-09-20 2008-07-17 The Johns Hopkins University Combinatorial therapy of cancer and infectious diseases with anti-B7-H1 antibodies
CN104945508B (zh) * 2007-06-18 2019-02-22 默沙东有限责任公司 针对人程序性死亡受体pd-1的抗体
WO2009111315A2 (fr) * 2008-02-29 2009-09-11 Mayo Foundation For Medical Education And Research Méthodes de réduction de l’inflammation granulomateuse
US8168757B2 (en) * 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
EP2350129B1 (fr) 2008-08-25 2015-06-10 Amplimmune, Inc. Compositions d'antagonistes de pd-1 et methodes d'utilisation associees
LT4209510T (lt) * 2008-12-09 2024-03-12 F. Hoffmann-La Roche Ag Anti-pd-l1 antikūnai ir jų panaudojimas t ląstelių funkcijos pagerinimui
WO2010102278A1 (fr) * 2009-03-06 2010-09-10 President And Fellows Of Harvard College Procédés et compositions permettant de générer et de maintenir des cellules t de régulation
US9783578B2 (en) 2010-06-25 2017-10-10 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
WO2014121085A1 (fr) 2013-01-31 2014-08-07 Thomas Jefferson University Protéines de fusion à base de pd-l1 et pd-l2 et leurs utilisations
US20160122829A1 (en) 2013-06-06 2016-05-05 Dana-Farber Cancer Institute, Inc. Compositions and Methods for Identification, Assessment, Prevention, and Treatment of Cancer Using PD-L1 Isoforms
BR112016005303A2 (pt) * 2013-09-11 2017-09-12 Medimmune Ltd anticorpos anti-b7-h1 para tratamento de tumores
TWI681969B (zh) 2014-01-23 2020-01-11 美商再生元醫藥公司 針對pd-1的人類抗體
TWI680138B (zh) 2014-01-23 2019-12-21 美商再生元醫藥公司 抗pd-l1之人類抗體
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
HUE052606T2 (hu) 2014-12-09 2021-05-28 Regeneron Pharma Humanizált differenciációs klaszter 274 génnel rendelkezõ nem humán állatok
MA41460A (fr) 2015-02-03 2017-12-12 Oncomed Pharm Inc Agents de liaison à la tnfrsf et leurs utilisations
KR20170132171A (ko) * 2015-02-26 2017-12-01 메르크 파텐트 게엠베하 암 치료를 위한 pd­1 / pd­l1 저해제
IL292449B2 (en) 2015-03-13 2024-02-01 Cytomx Therapeutics Inc Nucleic acids encoding antibodies against PDL1 and methods for their preparation
US11060150B2 (en) 2015-03-17 2021-07-13 Mayo Foundation For Medical Education And Research Methods and materials for assessing and treating cancer
KR20180018762A (ko) 2015-06-16 2018-02-21 메르크 파텐트 게엠베하 Pd-l1 길항제 조합 치료
AU2016306597A1 (en) 2015-08-07 2018-02-22 Pieris Pharmaceuticals Gmbh Novel fusion polypeptide specific for LAG-3 and PD-1
AR105654A1 (es) 2015-08-24 2017-10-25 Lilly Co Eli Anticuerpos pd-l1 (ligando 1 de muerte celular programada)
JP6654694B2 (ja) 2015-10-02 2020-02-26 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト 抗pd1抗体と使用方法
EP3365062A4 (fr) 2015-10-19 2019-07-17 Cold Genesys, Inc. Procédés de traitement de tumeurs solides ou lymphatiques par polythérapie
EP3426271A4 (fr) 2016-03-10 2019-10-16 Cold Genesys, Inc. Méthodes de traitement de tumeurs solides ou lymphatiques par polythérapie
AU2017238054B2 (en) 2016-03-21 2023-10-19 Dana-Farber Cancer Institute, Inc. T-cell exhaustion state-specific gene expression regulators and uses thereof
TWI822521B (zh) 2016-05-13 2023-11-11 美商再生元醫藥公司 藉由投予pd-1抑制劑治療皮膚癌之方法
US9567399B1 (en) * 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
KR20230044038A (ko) 2016-08-09 2023-03-31 키맵 리미티드 항-icos 항체
US11274154B2 (en) 2016-10-06 2022-03-15 Pfizer Inc. Dosing regimen of avelumab for the treatment of cancer
EP3534947A1 (fr) 2016-11-03 2019-09-11 Kymab Limited Anticorps, combinaisons comprenant des anticorps, biomarqueurs, utilisations et procédés
US10611823B2 (en) 2016-12-14 2020-04-07 Hanssen Biotech, Inc CD137 binding fibronectin type III domains
KR102568559B1 (ko) 2016-12-14 2023-08-18 얀센 바이오테크 인코포레이티드 Cd8a-결합 섬유결합소 iii형 도메인
US10597438B2 (en) 2016-12-14 2020-03-24 Janssen Biotech, Inc. PD-L1 binding fibronectin type III domains
WO2018134279A1 (fr) 2017-01-18 2018-07-26 Pieris Pharmaceuticals Gmbh Nouveaux polypeptides de fusion spécifiques de lag-3 et pd-1
MA47604A (fr) 2017-02-21 2020-01-01 Regeneron Pharma Anticorps anti-pd-1 pour le traitement du cancer du poumon
US11603407B2 (en) 2017-04-06 2023-03-14 Regeneron Pharmaceuticals, Inc. Stable antibody formulation
BR112019025188A2 (pt) 2017-06-01 2020-06-23 Cytomx Therapeutics, Inc. Anticorpos anti-pdl1 ativáveis e métodos de uso dos mesmos
JP2020523018A (ja) 2017-06-09 2020-08-06 プロビデンス ヘルス アンド サービシーズ−オレゴン がんの処置のための腫瘍反応性ヒトt細胞の同定のためのcd39およびcd103の使用
GB201709808D0 (en) 2017-06-20 2017-08-02 Kymab Ltd Antibodies
EP3728314A1 (fr) 2017-12-19 2020-10-28 Kymab Limited Anticorps bispécifique pour icos et pd-l1
JP7466459B2 (ja) 2018-04-17 2024-04-12 セルデックス セラピューティクス インコーポレイテッド 抗cd27および抗pd-l1抗体ならびに二重特異性構築物
US20210253614A1 (en) 2018-05-31 2021-08-19 Peloton Therapeutics, Inc. Compositions and methods for inhibiting cd73
WO2020021061A1 (fr) 2018-07-26 2020-01-30 Pieris Pharmaceuticals Gmbh Anticorps humanisés anti-pd-1 et leurs utilisations
CA3106881A1 (fr) 2018-08-27 2020-03-05 Pieris Pharmaceuticals Gmbh Polytherapies comprenant des agents bispecifiques cd137/her2 et des inhibiteurs de l'axe pd-1 et leurs utilisations
CA3117710A1 (fr) 2018-12-11 2020-06-18 Theravance Biopharma R&D Ip, Llc Inhibiteurs d'alk5
US20220107320A1 (en) 2019-02-15 2022-04-07 Incelldx, Inc. Assaying Bladder-Associated Samples, Identifying and Treating Bladder-Associated Neoplasia, and Kits for Use Therein
US11781138B2 (en) 2019-10-14 2023-10-10 Aro Biotherapeutics Company FN3 domain-siRNA conjugates and uses thereof
EP4045061A4 (fr) 2019-10-14 2024-04-17 Aro Biotherapeutics Company Domaines de type iii de fibronectine de liaison à cd137
CN114728941A (zh) 2019-11-22 2022-07-08 施万生物制药研发Ip有限责任公司 作为alk5抑制剂的经取代的1,5-萘啶或喹啉
TW202204339A (zh) 2020-03-31 2022-02-01 美商施萬生物製藥研發 Ip有限責任公司 經取代的嘧啶及使用方法
TW202214623A (zh) 2020-06-10 2022-04-16 美商施萬生物製藥研發 Ip有限責任公司 結晶型alk5抑制劑及其用途
WO2022251359A1 (fr) 2021-05-26 2022-12-01 Theravance Biopharma R&D Ip, Llc Inhibiteurs bicycliques de l'alk5 et procédés d'utilisation
EP4351595A1 (fr) 2021-06-07 2024-04-17 Providence Health & Services - Oregon Cxcr5, pd-1 et icos exprimant des lymphocytes t cd4 réactifs aux tumeurs et leur utilisation
CN117794953A (zh) 2021-08-03 2024-03-29 豪夫迈·罗氏有限公司 双特异性抗体及使用方法

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6641809B1 (en) * 1990-03-26 2003-11-04 Bristol-Myers Squibb Company Method of regulating cellular processes mediated by B7 and CD28
US5858776A (en) * 1993-11-03 1999-01-12 Repligen Corporation Tumor cells with increased immunogenicity and uses therefor
US6491925B2 (en) * 1996-08-15 2002-12-10 Emory University Compositions and methods for cancer prophylaxis and/or treatment
US7368531B2 (en) * 1997-03-07 2008-05-06 Human Genome Sciences, Inc. Human secreted proteins
US7411051B2 (en) 1997-03-07 2008-08-12 Human Genome Sciences, Inc. Antibodies to HDPPA04 polypeptide
US6562347B1 (en) * 1998-03-12 2003-05-13 The United States Of America As Represented By The Department Of Health And Human Services Chemokine-tumor antigen fusion proteins as cancer vaccines
US7041474B2 (en) * 1998-12-30 2006-05-09 Millennium Pharmaceuticals, Inc. Nucleic acid encoding human tango 509
US20080213778A1 (en) * 1998-12-30 2008-09-04 Millennium Pharmaceuticals, Inc. Novel genes encoding proteins having prognostic, diagnostic, preventive, therapeutic, and other uses
WO2001007611A2 (fr) 1999-07-26 2001-02-01 Genentech, Inc. Nouveaux polynucleotides et technique d'utilisation de ceux-ci
DE19939653A1 (de) * 1999-08-13 2001-02-22 Thomas Huenig Verwendung CD28 spezifischer monoklonaler Antikörper zur Herstellung einer pharmazeutischen Zusammensetzung
HU228477B1 (en) * 1999-08-23 2013-03-28 Dana Farber Cancer Inst Inc Pd-1, a receptor for b7-4, and uses therefor
PL362804A1 (en) * 1999-08-23 2004-11-02 Dana-Farber Cancer Institute Novel b7-4 molecules and uses therefor
US6803192B1 (en) * 1999-11-30 2004-10-12 Mayo Foundation For Medical Education And Research B7-H1, a novel immunoregulatory molecule
CA2392477A1 (fr) * 1999-11-30 2001-06-07 Mayo Foundation For Medical Education And Research Nouvelle molecule immunoregulatrice b7-h1,
US7029365B2 (en) * 2000-02-17 2006-04-18 Applied Materials Inc. Pad assembly for electrochemical mechanical processing
US20030031675A1 (en) * 2000-06-06 2003-02-13 Mikesell Glen E. B7-related nucleic acids and polypeptides useful for immunomodulation
AU2001275285A1 (en) * 2000-06-06 2001-12-17 Bristol-Myers Squibb Company B7-related nucleic acids and polypeptides and their uses for immunomodulation
AU7309601A (en) 2000-06-28 2002-01-08 Genetics Inst Pd-l2 molecules: novel pd-1 ligands and uses therefor
US6635750B1 (en) * 2000-07-20 2003-10-21 Millennium Pharmaceuticals, Inc. B7-H2 nucleic acids, members of the B7 family
AU2001283507A1 (en) * 2000-07-27 2002-02-13 Mayo Foundation For Medical Education And Research B7-h3 and b7-h4, novel immunoregulatory molecules
WO2002024891A2 (fr) * 2000-09-20 2002-03-28 Amgen, Inc. Molecules semblables a b7 (b7-like, b7-l) et utilisations correspondantes
MXPA03008959A (es) * 2001-04-02 2004-10-15 Wyeth Corp Pd-1, un receptor para b7-4 y sus usos.
AR036993A1 (es) * 2001-04-02 2004-10-20 Wyeth Corp Uso de agentes que modulan la interaccion entre pd-1 y sus ligandos en la submodulacion de respuestas inmunologicas
US7794710B2 (en) * 2001-04-20 2010-09-14 Mayo Foundation For Medical Education And Research Methods of enhancing T cell responsiveness
CA2466279A1 (fr) 2001-11-13 2003-05-22 Dana-Farber Cancer Institute, Inc. Agents modulant l'activite de cellules immunes et procedes d'utilisation associes
DE10212108A1 (de) * 2002-03-13 2003-10-02 Tegenero Ag Verwendung einer an CD28 bindenden Wirksubstanz zur Herstellung einer pharmazeutischen Zusammensetzung
US7432351B1 (en) * 2002-10-04 2008-10-07 Mayo Foundation For Medical Education And Research B7-H1 variants
US7449300B2 (en) * 2002-11-21 2008-11-11 Mayo Foundation For Medical Education And Research Detection of antibodies specific for B7-H1 in subjects with diseases or pathological conditions mediated by activated T cells
KR100518346B1 (ko) * 2003-12-01 2005-09-29 현대모비스 주식회사 스티어링 기어 일체형 프레임 구조
EP2889309B1 (fr) 2006-03-03 2017-12-27 Ono Pharmaceutical Co., Ltd. Multimère de domaine extracellulaire de pd-1 ou pd-l1

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BELL ELAINE: 'B7h and B7-H1: new members of the B7 family' IMMUNOLOGY TODAY vol. 21, no. 5, February 2000, page 59, XP004187227 *
DONG ET AL.: 'B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion' NATURE MEDICINE December 1999, pages 1365 - 1369, XP002156076 *
TAMURA ET AL.: 'B7-H1 costimulation preferentially enhances CD28-independent T-helper cell function' BLOOD vol. 97, no. 6, 15 March 2001, pages 1809 - 1816, XP002958254 *

Cited By (162)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8460927B2 (en) 1999-11-30 2013-06-11 Mayo Foundation For Medical Education And Research B7-H1 antibodies and method of use
EP2206517A1 (fr) * 2002-07-03 2010-07-14 Ono Pharmaceutical CO., LTD. Compositions immunostimulantes contenant un anticorps anti-PD-L1
US8728474B2 (en) 2002-07-03 2014-05-20 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
EP3287144A1 (fr) * 2002-07-03 2018-02-28 ONO Pharmaceutical Co., Ltd. Compositions immunostimulantes
US9067999B1 (en) 2002-07-03 2015-06-30 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US9073994B2 (en) 2002-07-03 2015-07-07 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US9439962B2 (en) 2002-07-03 2016-09-13 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US8168179B2 (en) 2002-07-03 2012-05-01 Ono Pharmaceutical Co., Ltd. Treatment method using anti-PD-L1 antibody
US9402899B2 (en) 2002-07-03 2016-08-02 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US9393301B2 (en) 2002-07-03 2016-07-19 Ono Pharmaceutical Co., Ltd. Immunopotentiative composition
US11242387B2 (en) 2004-10-06 2022-02-08 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US11939378B2 (en) 2004-10-06 2024-03-26 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US9803015B2 (en) 2004-10-06 2017-10-31 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US8747833B2 (en) 2004-10-06 2014-06-10 Mayo Foundation For Medical Education And Research B7-H1 and methods of diagnosis, prognosis, and treatment of cancer
US9387247B2 (en) 2005-05-09 2016-07-12 Ono Pharmaceutical Co., Ltd. Monoclonal antibodies to programmed death 1 (PD-1)
US9492540B2 (en) 2005-05-09 2016-11-15 Ono Pharmaceutical Co., Ltd. Methods for treating cancer using anti-PD-1 antibodies
US8779105B2 (en) 2005-05-09 2014-07-15 Medarex, L.L.C. Monoclonal antibodies to programmed death 1 (PD-1)
US9084776B2 (en) 2005-05-09 2015-07-21 E.R. Squibb & Sons, L.L.C. Methods for treating cancer using anti-PD-1 antibodies
US9358289B2 (en) 2005-05-09 2016-06-07 Ono Pharmaceutical Co., Ltd. Methods for treating cancer using anti-PD-1 antibodies in combination with anti-CTLA-4 antibodies
US9492539B2 (en) 2005-05-09 2016-11-15 Ono Pharmaceutical Co., Ltd. Monoclonal antibodies to Programmed Death 1 (PD-1)
US10441655B2 (en) 2005-05-09 2019-10-15 Ono Pharmaceutical Co., Ltd. Monoclonal antibodies to programmed death 1 (PD-1)
US11359013B2 (en) 2005-06-08 2022-06-14 Emory University Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (PD-1) pathway
US10370446B2 (en) 2005-06-08 2019-08-06 Emory University Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (PD-1) pathway
EP2397156B1 (fr) 2005-06-08 2016-11-02 Dana-Farber Cancer Institute, Inc. Procédés et compositions pour le traitement du cancer et d'infections pérsistantes en inhibant la voie de la mort céllulaire programmée (PD-1)
US9457080B2 (en) 2005-06-08 2016-10-04 Emory University Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (PD-1) pathway
US8652465B2 (en) 2005-06-08 2014-02-18 Emory University Methods and compositions for the treatment of persistent infections
EP2397155B1 (fr) 2005-06-08 2017-12-13 Dana-Farber Cancer Institute, Inc. Procédés et compositions pour le traitement d'infections persistantes en inhibant la voie de la mort cellulaire programmée (PD-1)
EP1895842A4 (fr) * 2005-06-17 2009-06-03 Univ Rochester Procedes et compositions permettant d'ameliorer la memoire immune par blocage de l'effacement intrahepatique des lymphocytes t actives
EP1895842A2 (fr) * 2005-06-17 2008-03-12 University of Rochester Procedes et compositions permettant d'ameliorer la memoire immune par blocage de l'effacement intrahepatique des lymphocytes t actives
US9273135B2 (en) 2005-07-01 2016-03-01 E. R. Squibb & Sons, L. L. C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US20160075782A1 (en) 2005-07-01 2016-03-17 E.R. Squibb & Sons, L. L. C. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US9580507B2 (en) 2005-07-01 2017-02-28 E.R. Squibb & Sons, L. L. C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US9580505B2 (en) 2005-07-01 2017-02-28 E.R. Squibb & Sons, L. L. C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US9102725B2 (en) 2005-07-01 2015-08-11 E. R. Squibb & Sons, L. L. C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US8383796B2 (en) 2005-07-01 2013-02-26 Medarex, Inc. Nucleic acids encoding monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2007100098A1 (fr) 2006-03-03 2007-09-07 Kyoto University Multimere de domaine extracellulaire d'une molecule fonctionnelle de surface cellulaire
EP2889309A1 (fr) 2006-03-03 2015-07-01 Ono Pharmaceutical Co., Ltd. Tétramère de domaine extracellulaire de PD-L1
EP1997887A4 (fr) * 2006-03-03 2009-06-03 Univ Kyoto Multimere de domaine extracellulaire d'une molecule fonctionnelle de surface cellulaire
EP1997887A1 (fr) * 2006-03-03 2008-12-03 Kyoto University Multimere de domaine extracellulaire d'une molecule fonctionnelle de surface cellulaire
US8574872B2 (en) 2006-03-03 2013-11-05 Ono Pharmaceutical Co., Ltd. Multimer of extracellular domain of cell surface functional molecule
US8216996B2 (en) 2006-03-03 2012-07-10 Ono Pharmaceutical Co., Ltd. Multimer of extracellular domain of cell surface functional molecule
EP2468765A1 (fr) 2006-03-03 2012-06-27 ONO Pharmaceutical Co., Ltd. Multimère de domaine extracellulaire d'une molécule fonctionnelle de surface cellulaire
JP2017002077A (ja) * 2006-12-27 2017-01-05 エモリー ユニバーシティ 感染症および腫瘍を処置するための組成物および方法
EP2133365B1 (fr) 2006-12-27 2017-05-17 Emory University Compositions et procédés pour le traitement d'infections et de tumeurs
WO2009089149A1 (fr) * 2008-01-03 2009-07-16 The Johns Hopkins University Antagonistes de b7-h1 (cd274) induisant l'apoptose de cellules tumorales
AU2009288289B2 (en) * 2008-08-25 2012-11-08 Amplimmune, Inc. PD-1 antagonists and methods of use thereof
WO2010098788A2 (fr) * 2008-08-25 2010-09-02 Amplimmune, Inc. Antagonistes de pd-i et procédés de traitement d'une maladie infectieuse
WO2010098788A3 (fr) * 2008-08-25 2010-12-02 Amplimmune, Inc. Antagonistes de pd-i et procédés de traitement d'une maladie infectieuse
US20120201824A1 (en) * 2008-11-11 2012-08-09 Mariusz Wasik Compositions and Methods for Inhibiting an Oncogenic Protein to Enhance Immunogenicity
US9598491B2 (en) 2008-11-28 2017-03-21 Emory University Methods for the treatment of infections and tumors
WO2011161699A2 (fr) 2010-06-25 2011-12-29 Aurigene Discovery Technologies Limited Composés modulateurs de l'immunosuppression
WO2012145493A1 (fr) 2011-04-20 2012-10-26 Amplimmune, Inc. Anticorps et autres molécules qui se lient à b7-h1 et à pd-1
US9205148B2 (en) 2011-04-20 2015-12-08 Medimmune, Llc Antibodies and other molecules that bind B7-H1 and PD-1
EP3403672A1 (fr) 2011-04-20 2018-11-21 Medlmmune, LLC Anticorps et autres molécules se liant à b7-h1 et pd-1
US9096642B2 (en) 2011-06-08 2015-08-04 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
WO2012168944A1 (fr) 2011-06-08 2012-12-13 Aurigene Discovery Technologies Limited Composés thérapeutiques pour une immunomodulation
US9669078B2 (en) 2011-10-17 2017-06-06 Herlev Hospital PD-L1 based immunotherapy
WO2013056716A1 (fr) * 2011-10-17 2013-04-25 Herlev Hospital Immunothérapie par pd-l1
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
US11884724B2 (en) 2011-11-28 2024-01-30 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
EP3763741A1 (fr) 2011-11-28 2021-01-13 Merck Patent GmbH Anticorps anti-pd-l1 et leurs utilisations
US10759856B2 (en) 2011-11-28 2020-09-01 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
US9624298B2 (en) 2011-11-28 2017-04-18 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
US10487147B2 (en) 2011-11-28 2019-11-26 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
WO2013132317A1 (fr) 2012-03-07 2013-09-12 Aurigene Discovery Technologies Limited Composés peptidomimétiques utilisés comme immunomodulateurs
US9422339B2 (en) 2012-03-29 2016-08-23 Aurigene Discovery Technologies Limited Immunomodulating cyclic compounds
WO2013144704A1 (fr) 2012-03-29 2013-10-03 Aurigene Discovery Technologies Limited Composés cycliques d'immunomodulation provenant de la boucle bc de pd1 humain
US9856320B2 (en) 2012-05-15 2018-01-02 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10072082B2 (en) 2012-05-15 2018-09-11 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US9212224B2 (en) 2012-05-15 2015-12-15 Bristol-Myers Squibb Company Antibodies that bind PD-L1 and uses thereof
US10266596B1 (en) 2012-05-15 2019-04-23 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10577423B2 (en) 2012-05-15 2020-03-03 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10584170B2 (en) 2012-05-15 2020-03-10 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10323093B2 (en) 2012-05-15 2019-06-18 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10323092B2 (en) 2012-05-15 2019-06-18 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10316091B2 (en) 2012-05-15 2019-06-11 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10266595B2 (en) 2012-05-15 2019-04-23 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10316090B2 (en) 2012-05-15 2019-06-11 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10604575B2 (en) 2012-05-15 2020-03-31 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10308714B2 (en) 2012-05-15 2019-06-04 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10266594B1 (en) 2012-05-15 2019-04-23 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
US10138299B2 (en) 2012-05-15 2018-11-27 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
EP3292873B1 (fr) 2013-02-22 2019-05-01 CureVac AG Combinaison de vaccination et d'inhibition de la voie pd-1
US11458195B2 (en) 2013-02-22 2022-10-04 Curevac Ag Combination of vaccination and inhibition of the PD-1 pathway
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
WO2014194293A1 (fr) 2013-05-30 2014-12-04 Amplimmune, Inc. Méthodes améliorées de sélection de patients pouvant être soumis à des thérapies ciblant pd-1 ou b7-h4, et polythérapies associées
US10106581B2 (en) 2013-09-06 2018-10-23 Aurigene Discovery Technologies Limited Cyclic peptidomimetic compounds as immunomodulators
US9771338B2 (en) 2013-09-06 2017-09-26 Auirgene Discovery Technologies Limited 1,2,4-oxadiazole derivatives as immunomodulators
US10961205B2 (en) 2013-09-06 2021-03-30 Aurigene Discovery Technologies Limited 1,2,4-oxadiazole derivatives as immunomodulators
WO2015033303A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Composés peptidomimétiques cycliques utilisés comme immunomodulateurs
US10173989B2 (en) 2013-09-06 2019-01-08 Aurigene Discovery Technologies Limited 1,2,4-oxadiazole derivatives as immunomodulators
US10160736B2 (en) 2013-09-06 2018-12-25 Aurigene Discovery Technologies Limited 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
US10590093B2 (en) 2013-09-06 2020-03-17 Aurigene Discovery Technologies Limited 1,2,4-oxadiazole derivatives as immunomodulators
US9776978B2 (en) 2013-09-06 2017-10-03 Aurigene Discovery Technologies Limited 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
WO2015033299A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,2,4-oxadiazole utilisés comme immunomodulateurs
EP3385257A1 (fr) 2013-09-06 2018-10-10 Aurigene Discovery Technologies Limited Dérivés 1,3,4-oxadiazole et 1,3,4-thiadiazole servant d'immunomodulateurs
EP3366289A1 (fr) 2013-09-06 2018-08-29 Aurigene Discovery Technologies Limited Composés peptidomimétiques cycliques utilisés comme immunomodulateurs
EP3363790A1 (fr) 2013-09-06 2018-08-22 Aurigene Discovery Technologies Limited Dérivés 1,2,4-oxadiazole utilisés comme immunomodulateurs
WO2015033301A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,3,4-oxadiazole et 1,3,4-thiadiazole servant d'immunomodulateurs
US11512060B2 (en) 2013-09-06 2022-11-29 Aurigene Discovery Technologies Limited 1,2,4-oxadiazole derivatives as immunomodulators
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
US11708412B2 (en) 2013-09-26 2023-07-25 Novartis Ag Methods for treating hematologic cancers
US11136393B2 (en) 2013-10-01 2021-10-05 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of Bim
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US9815898B2 (en) 2014-01-24 2017-11-14 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
US11827704B2 (en) 2014-01-24 2023-11-28 Novartis Ag Antibody molecules to PD-1 and uses thereof
US9683048B2 (en) 2014-01-24 2017-06-20 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US10981990B2 (en) 2014-01-31 2021-04-20 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11155620B2 (en) 2014-01-31 2021-10-26 Novartis Ag Method of detecting TIM-3 using antibody molecules to TIM-3
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US11098119B2 (en) 2014-06-26 2021-08-24 Macrogenics, Inc. Covalently bonded diabodies having immunoreactivity with PD-1 and LAG-3, and methods of use thereof
US10160806B2 (en) 2014-06-26 2018-12-25 Macrogenics, Inc. Covalently bonded diabodies having immunoreactivity with PD-1 and LAG-3, and methods of use thereof
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
US11504376B2 (en) 2014-07-23 2022-11-22 Mayo Foundation For Medical Education And Research Targeting DNA-PKCS and B7-H1 to treat cancer
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
US9988452B2 (en) 2014-10-14 2018-06-05 Novartis Ag Antibody molecules to PD-L1 and uses thereof
US10851165B2 (en) 2014-10-14 2020-12-01 Novartis Ag Antibody molecules to PD-L1 and methods of treating cancer
WO2016142833A1 (fr) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited Composés 1,2,4-oxadiazoles et thiadiazoles utilisés comme immunomodulateurs
EP4023645A1 (fr) 2015-03-10 2022-07-06 Aurigene Discovery Technologies Limited Composés 1,2,4-oxadiazoles et thiadiazoles utilisés comme immunomodulateurs
US11465976B2 (en) 2015-03-10 2022-10-11 Aurigene Discovery Technologies Limited 1,2,4-oxadiazole and thiadiazole compounds as immunomodulators
US10781189B2 (en) 2015-03-10 2020-09-22 Aurigene Discovery Technologies Limited 1,2,4-Oxadiazole and thiadiazole compounds as immunomodulators
US11078279B2 (en) 2015-06-12 2021-08-03 Macrogenics, Inc. Combination therapy for the treatment of cancer
WO2017009842A2 (fr) 2015-07-16 2017-01-19 Biokine Therapeutics Ltd. Compositions et méthodes pour le traitement du cancer
EP3744340A2 (fr) 2015-07-16 2020-12-02 Biokine Therapeutics Ltd. Compositions et procédés pour le traitement du cancer
EP3943098A2 (fr) 2015-07-16 2022-01-26 Biokine Therapeutics Ltd. Compositions et procédés pour le traitement du cancer
US11623959B2 (en) 2015-07-30 2023-04-11 Macrogenics, Inc. PD-1-binding molecules and methods of use thereof
EP3456346A1 (fr) 2015-07-30 2019-03-20 MacroGenics, Inc. Molécules de liaison pd-1 et lag-3 et leurs procédés d'utilisation
US10577422B2 (en) 2015-07-30 2020-03-03 Macrogenics, Inc. PD-1-binding molecules and methods of use thereof
EP3981792A1 (fr) 2015-07-30 2022-04-13 MacroGenics, Inc. Molécules de liaison pd-1 et leurs procédés d'utilisation
US11174315B2 (en) 2015-10-08 2021-11-16 Macrogenics, Inc. Combination therapy for the treatment of cancer
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1
WO2017106061A1 (fr) 2015-12-14 2017-06-22 Macrogenics, Inc. Molécules bispécifiques présentant une immunoréactivité par rapport à pd-1 et à ctla-4 et leurs procédés d'utilisation
US10954301B2 (en) 2015-12-14 2021-03-23 Macrogenics, Inc. Bispecific molecules having immunoreactivity with PD-1 and CTLA-4, and methods of use thereof
US11840571B2 (en) 2015-12-14 2023-12-12 Macrogenics, Inc. Methods of using bispecific molecules having immunoreactivity with PD-1 and CTLA-4
WO2017106372A1 (fr) 2015-12-15 2017-06-22 Oncoimmune, Inc. Anticorps monoclonaux chimériques et humanisés anti-ctla4 humaine, et leurs utilisations
WO2017112741A1 (fr) 2015-12-22 2017-06-29 Novartis Ag Récepteur d'antigène chimérique (car) contre la mésothéline et anticorps contre l'inhibiteur de pd-l1 pour une utilisation combinée dans une thérapie anticancéreuse
WO2017203362A1 (fr) * 2016-05-25 2017-11-30 The Council Of The Queensland Institute Of Medical Research Inhibiteurs du point de contrôle immunitaire et lymphocytes t cytotoxiques pour le traitement du cancer
CN109496155A (zh) * 2016-05-25 2019-03-19 昆士兰医学研究所理事会 用于治疗癌症的免疫检查点抑制剂和细胞毒性t细胞
US11459394B2 (en) 2017-02-24 2022-10-04 Macrogenics, Inc. Bispecific binding molecules that are capable of binding CD137 and tumor antigens, and uses thereof
US11942149B2 (en) 2017-02-24 2024-03-26 Macrogenics, Inc. Bispecific binding molecules that are capable of binding CD137 and tumor antigens, and uses thereof
US11236167B2 (en) 2017-04-17 2022-02-01 Joint Stock Company “Biocad” Monoclonal antibody to PD-L1
WO2018194496A2 (fr) 2017-04-17 2018-10-25 Закрытое Акционерное Общество "Биокад" Anticorps monoclonal à pd-l1
US11643401B2 (en) 2017-09-29 2023-05-09 Curis, Inc. Crystal forms of immunomodulators
US11040948B2 (en) 2017-09-29 2021-06-22 Curis, Inc. Crystal forms of immunomodulators
US11939306B2 (en) 2017-09-29 2024-03-26 Curis, Inc. Crystal forms of immunomodulators
US11840567B2 (en) 2017-10-03 2023-12-12 Joint Stock Company “Biocad” Bispecific antibodies with specific binding to CD47 and PD-L1
US11136300B2 (en) 2017-10-11 2021-10-05 Aurigene Discovery Technologies Limited Crystalline forms of 3-substituted 1,2,4-oxadiazole
US11680051B2 (en) 2017-10-11 2023-06-20 Aurigene Discovery Technologies Limited Crystalline forms of 3-substituted 1,2,4-oxadiazole
US11497734B2 (en) 2017-11-03 2022-11-15 Aurigene Discovery Technologies Limited Dual inhibitors of TIM-3 and PD-1 pathways
US11497735B2 (en) 2017-11-06 2022-11-15 Aurigene Discovery Technologies Limited Conjoint therapies for immunomodulation
US11673894B2 (en) 2018-02-27 2023-06-13 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as A2A / A2B inhibitors
US11873304B2 (en) 2018-05-18 2024-01-16 Incyte Corporation Fused pyrimidine derivatives as A2A/A2B inhibitors
WO2019246110A1 (fr) 2018-06-20 2019-12-26 Incyte Corporation Anticorps anti-pd-1 et leurs utilisations
EP4349411A2 (fr) 2018-06-20 2024-04-10 Incyte Corporation Anticorps anti-pd-1 et leurs utilisations
WO2020132560A2 (fr) 2018-12-21 2020-06-25 Aim Immunotech Inc. Compositions et méthodes pour cancérothérapie
US11884665B2 (en) 2019-01-29 2024-01-30 Incyte Corporation Pyrazolopyridines and triazolopyridines as A2A / A2B inhibitors
WO2021138512A1 (fr) 2020-01-03 2021-07-08 Incyte Corporation Polythérapie comprenant des inhibiteurs d'a2a/a2b et de pd-1/pd-l1
WO2021158635A1 (fr) 2020-02-07 2021-08-12 Al Therapeutics, Inc. Compositions antivirales et procédés d'utilisation
WO2022147092A1 (fr) 2020-12-29 2022-07-07 Incyte Corporation Polythérapie comprenant des inhibiteurs a2a/a2b, des inhibiteurs pd-1/pd-l1 et des anticorps anti-cd73

Also Published As

Publication number Publication date
AU2002258941A1 (en) 2002-11-05
WO2002086083A3 (fr) 2003-02-27
US20030039653A1 (en) 2003-02-27
US20120027759A1 (en) 2012-02-02
US7794710B2 (en) 2010-09-14

Similar Documents

Publication Publication Date Title
US7794710B2 (en) Methods of enhancing T cell responsiveness
AU2019203823B2 (en) CS1-specific chimeric antigen receptor engineered immune effector cells
US8772026B2 (en) Enhancement of immune responses by 4-1 BB-binding agents
US8460927B2 (en) B7-H1 antibodies and method of use
Baxevanis et al. Cancer immunotherapy
US8207130B2 (en) Combination cancer immunotherapy with co-stimulatory molecules
Weinberg et al. Engagement of the OX-40 receptor in vivo enhances antitumor immunity
van den Broeke et al. Dendritic cell-induced activation of adaptive and innate antitumor immunity
US20170204185A1 (en) B7-h3 and b7-h4, novel immunoregulatory molecules
Tutt et al. T cell immunity to lymphoma following treatment with anti-CD40 monoclonal antibody
JP2021513839A (ja) 改変された多能性幹細胞並びに製造方法及び使用方法
JP2002504334A (ja) Ox−40レセプター結合因子又はそれをコードする核酸を含む組成物並びに抗原特異的免疫応答を増強するための方法
TW202012436A (zh) 嵌合hla輔助(accessory)受體
WO2012015662A1 (fr) Récepteur de l'il-18 en tant que nouvelle cible des lymphocytes t régulateurs dans le cancer
Mansoor et al. Genetically Modified T Cell Therapy Optimisation of the Chimeric T Cell Receptor for Cancer Therapy

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP